1
|
Gintoni I, Mastrogeorgiou M, Papakosta V, Vassiliou S, Yapijakis C. Genetic Variations Related to Angiotensin II Production and Risk for Basal Cell Carcinoma. Biochem Genet 2025; 63:917-935. [PMID: 38546913 DOI: 10.1007/s10528-024-10746-0] [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: 05/30/2023] [Accepted: 02/16/2024] [Indexed: 03/23/2025]
Abstract
Basal cell carcinoma (BCC) is the most prevalent human neoplasm, with constantly increasing annual incidence. Despite its slow growth, BCC is locally invasive and, if left untreated, can cause severe complications, including metastasis and death. The renin-angiotensin system (RAS) plays a key role in electrolyte balance, atrial pressure, tissue development, homeostasis, and inflammation, but also in cancer development. After binding to its type 1 receptor (AT1R), angiotensin II (ANGII), the system's principal hormonal effector, regulates cancer pathways spanning from the formation of the initial cancer cell to the construction and nutrition of the tumor microenvironment, angiogenesis, proliferation, and metastasis. Although the role of RAS in the development of skin pathologies has not been widely researched, RAS-targeting antihypertensive medications have been shown to have a chemoprotective effect against BCC. Based on those findings, our group conducted a series of genetic association studies to investigate the association between common functional variations in key genes related to ANGII production (AGT, ACE, ACE2, AT1R, AT2R, and CMA1) and the risk of BCC occurrence. This review provides a summary of the current understanding of the ANGII involvement in BCC development. The reliable and easily assessed pool of genetic biomarkers may be used for predictive testing and prevention purposes in high-risk individuals.
Collapse
Affiliation(s)
- Iphigenia Gintoni
- Unit of Orofacial Genetics, 1st Department of Pediatrics, National Kapodistrian University of Athens, University Research Institute for the Study of Genetic and Malignant Disorders in Childhood, Choremion Laboratory "Hagia Sophia" Children's Hospital, Athens, Greece
- Department of Molecular Genetics, Cephalogenetics Center, Philaretou 88, Kallithea, 17675, Athens, Greece
- Department of Oral and Maxillofacial Surgery, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, Athens, Greece
| | - Michael Mastrogeorgiou
- Unit of Orofacial Genetics, 1st Department of Pediatrics, National Kapodistrian University of Athens, University Research Institute for the Study of Genetic and Malignant Disorders in Childhood, Choremion Laboratory "Hagia Sophia" Children's Hospital, Athens, Greece
| | - Veronica Papakosta
- Department of Oral and Maxillofacial Surgery, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, Athens, Greece
| | - Stavros Vassiliou
- Department of Oral and Maxillofacial Surgery, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, Athens, Greece
| | - Christos Yapijakis
- Unit of Orofacial Genetics, 1st Department of Pediatrics, National Kapodistrian University of Athens, University Research Institute for the Study of Genetic and Malignant Disorders in Childhood, Choremion Laboratory "Hagia Sophia" Children's Hospital, Athens, Greece.
- Department of Molecular Genetics, Cephalogenetics Center, Philaretou 88, Kallithea, 17675, Athens, Greece.
- Department of Oral and Maxillofacial Surgery, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, Athens, Greece.
| |
Collapse
|
2
|
Elsaafien K, Kirchner MK, Scott KA, Spector EA, Mowry FE, Sumners C, Stern JE, de Kloet AD, Krause EG. Neurons of the Central Nucleus of the Amygdala That Express Angiotensin Type 2 Receptors Couple Lowered Blood Pressure with Anxiolysis in Male Mice. J Neurosci 2025; 45:e1482242025. [PMID: 39909561 PMCID: PMC11924993 DOI: 10.1523/jneurosci.1482-24.2025] [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: 08/05/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
Relief from psychological stress confers cardio-protection by altering brain activity and lowering blood pressure; however, the neuronal circuits orchestrating these effects are unknown. Here, we used male mice to discern neuronal circuits conferring stress relief and reduced blood pressure. We found that neurons residing in the central nucleus of the amygdala (CeA) expressing angiotensin type 2 receptors (AT2R), deemed CeAAT2R, innervate brain nuclei regulating stress responding. In vivo optogenetic excitation of CeAAT2R lowered blood pressure, and this effect was abrogated by systemic hexamethonium or antagonism of GABA receptors within the CeA. Intriguingly, in vivo optogenetic excitation of CeAAT2R was also potently anxiolytic. Delivery of an AT2R agonist into the CeA recapitulated the hypotensive and anxiolytic effects, but ablating AT2R(s) from the CeA was anxiogenic. The results suggest that the excitation of CeAAT2R couples lowered blood pressure with anxiolysis. The implication is that therapeutics targeting CeAAT2R may provide stress relief and protection against cardiovascular disease.
Collapse
Affiliation(s)
- Khalid Elsaafien
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, Georgia 30303
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences Georgia State University, Atlanta, Georgia 30303
| | - Matthew K Kirchner
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, Georgia 30303
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences Georgia State University, Atlanta, Georgia 30303
| | - Karen A Scott
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, Georgia 30303
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences Georgia State University, Atlanta, Georgia 30303
| | - Eliot A Spector
- Departments of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610
| | - Francesca E Mowry
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, Georgia 30303
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences Georgia State University, Atlanta, Georgia 30303
| | - Colin Sumners
- Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Javier E Stern
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, Georgia 30303
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences Georgia State University, Atlanta, Georgia 30303
| | - Annette D de Kloet
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, Georgia 30303
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences Georgia State University, Atlanta, Georgia 30303
| | - Eric G Krause
- Neuroscience Institute, College of Arts and Sciences, Georgia State University, Atlanta, Georgia 30303
- Center for Neuroinflammation and Cardiometabolic Diseases, College of Arts and Sciences Georgia State University, Atlanta, Georgia 30303
| |
Collapse
|
3
|
Islam MA, Ford Versypt AN. Mathematical modeling of impacts of patient differences on renin-angiotensin system and applications to COVID-19 lung fibrosis outcomes. Comput Biol Med 2025; 186:109631. [PMID: 39753028 PMCID: PMC11932320 DOI: 10.1016/j.compbiomed.2024.109631] [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: 08/10/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 02/20/2025]
Abstract
Patient-specific premorbidity, age, and sex are significant heterogeneous factors that influence the severe manifestation of lung diseases, including COVID-19 fibrosis. The renin-angiotensin system (RAS) plays a prominent role in regulating the effects of these factors. Recent evidence shows patient-specific alterations of RAS peptide homeostasis concentrations with premorbidity and the expression level of angiotensin-converting enzyme 2 (ACE2) during COVID-19. However, conflicting evidence suggests decreases, increases, or no changes in RAS peptides after SARS-CoV-2 infection. A multiscale computational model was developed to quantify the systemic contribution of heterogeneous factors of RAS during COVID-19. Three submodels were connected-an agent-based model for in-host COVID-19 response in the lung tissue, a RAS dynamics model, and a fibrosis dynamics model to investigate the effects of patient-group-specific factors in the systemic alteration of RAS and collagen deposition in the lung. The model results indicated cell death due to inflammatory response as a major contributor to the reduction of ACE and ACE2. The model explained possible mechanisms for conflicting evidence of patient-group-specific changes in RAS peptides in previously published studies. RAS peptides decreased for all virtual patient groups with aging in both sexes. In contrast, large variations in the magnitude of reduction were observed between male and female virtual patients in the older and middle-aged groups. The patient-specific variations in homeostasis RAS peptide concentrations of SARS-CoV-2-negative patients affected the dynamics of RAS during infection. This model may find further applications in patient-specific calibrations of tissue models for acute and chronic lung diseases to develop personalized treatments.
Collapse
Affiliation(s)
- Mohammad Aminul Islam
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Ashlee N Ford Versypt
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA; Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA; Institute for Artificial Intelligence and Data Science, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA; Witebsky Center for Microbial Pathogenesis and Immunology, University at Buffalo, The State University of New York, Buffalo, NY, 14203, USA; Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14215, USA.
| |
Collapse
|
4
|
Leinweber ME, Walter C, Assadian A, Kopecky C, Domenig O, Kovarik JJ, Hofmann AG. Angiotensin Dysregulation in Patients with Arterial Aneurysms. Int J Mol Sci 2025; 26:1502. [PMID: 40003968 PMCID: PMC11855860 DOI: 10.3390/ijms26041502] [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: 01/11/2025] [Revised: 02/01/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Besides playing a critical role in maintaining cardiovascular homeostasis, the renin-angiotensin-aldosterone system (RAS) has been strongly implicated in (aortic) aneurysm pathogenesis. This study aims to investigate systemic and local levels of angiotensin (Ang) and its metabolites in patients with arterial aneurysms, predominantly abdominal aortic aneurysms, using advanced biochemical profiling techniques to provide new insights into the involvement of RAS in aneurysm genesis. A prospective, single-center study was conducted between October 2023 and July 2024. Serum Ang metabolite levels were measured using RAS Fingerprint technology. Aortic tissue samples were analyzed for local RAS activity, including Ang levels and enzyme activity. Additionally, pre- and postoperative serum samples were obtained in a select group of patients. In total, 37 aneurysm patients and 56 controls were included. Aneurysm patients exhibited higher systemic levels of nearly all Ang metabolites compared to controls, with significant differences in Ang I (p = 0.002), Ang II (p = 0.047), Ang 1-5 (p = 0.004), and Renin (p = 0.014) in patients without pharmacological RAS interference. Aneurysm patients receiving ACE inhibitors showed lower serum concentrations in ACE2 activity (p = 0.042) and increased Ang IV levels (p = 0.049) compared to controls. Postoperative measurements indicated different dynamics regarding angiotensin metabolite changes in patients with or without ACE inhibition. This study provides the first comprehensive characterization of RAS profiles in aneurysm patients. These findings add to the body of evidence regarding associations between of RAS and the pathogenesis of arterial aneurysms.
Collapse
Affiliation(s)
| | - Corinna Walter
- Department of Vascular and Endovascular Surgery, Clinic Ottakring, 1160 Vienna, Austria
| | - Afshin Assadian
- Department of Vascular and Endovascular Surgery, Clinic Ottakring, 1160 Vienna, Austria
| | - Chantal Kopecky
- Clinical Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Johannes Josef Kovarik
- Clinical Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Amun Georg Hofmann
- Department of Vascular and Endovascular Surgery, Clinic Ottakring, 1160 Vienna, Austria
| |
Collapse
|
5
|
Pathak A, Agrawal DK. Role of Gut Microbiota in Long COVID: Impact on Immune Function and Organ System Health. ARCHIVES OF MICROBIOLOGY & IMMUNOLOGY 2025; 9:38-53. [PMID: 40051430 PMCID: PMC11883900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/09/2025]
Abstract
SARS-CoV-2 infection has led to a range of long-lasting symptoms, collectively referred to as long COVID. Current research highlights the critical role of angiotensin-converting enzyme 2 (ACE2) in regulating gut microbiota diversity, vascular function, and homeostasis within the renin-angiotensin system (RAS). ACE2 is utilized by the SARS-CoV-2 virus to enter host cells, but its downregulation following infection contributes to gut microbiota dysbiosis and RAS disruption. These imbalances have been linked to a range of long COVID symptoms, including joint pain, chest pain, chronic cough, fatigue, brain fog, anxiety, depression, myalgia, peripheral neuropathy, memory difficulties, and impaired attention. This review investigates the dysregulation caused by SARS-CoV-2 infection and the long-term effects it has on various organ systems, including the musculoskeletal, neurological, renal, respiratory, and cardiovascular systems. We explored the bidirectional interactions between the gut microbiota, immune function, and these organ systems, focusing on how microbiota dysregulation contributes to the chronic inflammation and dysfunction observed in long COVID symptoms. Understanding these interactions is key for identifying effective therapeutic strategies and interventional targets aimed at mitigating the impact of long COVID on organ health and improving patient outcomes.
Collapse
Affiliation(s)
- Angelie Pathak
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766 USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766 USA
| |
Collapse
|
6
|
Huang JH, Lourenço BN, Coleman AE. The renin-angiotensin-aldosterone system in kidney diseases of cats and dogs. Vet J 2025; 309:106287. [PMID: 39672318 DOI: 10.1016/j.tvjl.2024.106287] [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: 08/27/2024] [Revised: 12/04/2024] [Accepted: 12/04/2024] [Indexed: 12/15/2024]
Abstract
The renin-angiotensin-aldosterone system (RAAS) has a well-established key pathophysiologic role in kidney diseases, and pharmacotherapy targeting this system is a mainstay of treatment of affected human beings, cats, and dogs. Several studies have evaluated the circulating RAAS in animals with spontaneous or experimentally induced kidney diseases. Evidence supporting the activation of this system has been demonstrated in some - but not all - studies and individuals, and the interindividual variability in circulating RAAS markers is high. Advances over the last few decades have expanded our understanding of the system, which now includes the existence of a counterbalancing "alternative" RAAS and tissular renin-angiotensin systems (RASs), the latter regulated independently of the circulating endocrine RAAS. The local RAS in the kidney, termed the intrarenal RAS, is currently recognized as an important regulator of kidney function and mediator of kidney disease. In general, information on the intrarenal RAS is lacking in cats and dogs with kidney diseases; however, existing limited data suggest its activation. Despite the inconsistent evidence for circulating RAAS activation in chronic kidney diseases, RAAS inhibitors have proven effective for the treatment of its common comorbidities, systemic arterial hypertension and renal proteinuria, in both cats and dogs. Further research of the circulating RAAS, the intrarenal RAS, and the interplay between these systems in the context of kidney diseases in companion animals might contribute to the development or refinement of future treatment strategies.
Collapse
Affiliation(s)
- Jane Hc Huang
- Department of Small Animal Medicine and Surgery, University of Georgia, College of Veterinary Medicine, Athens 30601, USA
| | - Bianca N Lourenço
- Department of Small Animal Medicine and Surgery, University of Georgia, College of Veterinary Medicine, Athens 30601, USA.
| | - Amanda E Coleman
- Department of Small Animal Medicine and Surgery, University of Georgia, College of Veterinary Medicine, Athens 30601, USA
| |
Collapse
|
7
|
Zhu F, Wang Z, Davis K, McSwiggin H, Zyuzin J, Liu J, Yan W, Rehan VK, Jendzjowsky N. Epigenetic Upregulation of Carotid Body Angiotensin Signaling Increases Blood Pressure. Hypertension 2025; 82:293-305. [PMID: 39633580 PMCID: PMC11732265 DOI: 10.1161/hypertensionaha.124.23349] [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: 05/14/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Epigenetic changes can be shaped by a wide array of environmental cues, maternal health, and behaviors. One of the most detrimental behaviors to the developing fetus is nicotine exposure. Perinatal nicotine exposure remains a significant risk factor for cardiovascular health and, in particular, hypertension. Increased basal carotid body (CB) activity and excitation are significant contributors to hypertension. This study investigated the epigenetic changes to CB activity induced by perinatal nicotine exposure resulting in CB-mediated hypertension. METHODS We used a rodent model of perinatal nicotine exposure and cell culture methods. RESULTS We show that the AgtR1 (angiotensin II type 1 receptor) is upregulated in the carotid bodies of nicotine-exposed offspring. These changes were attributed to an upregulation of genetic promotion as DNA methylation of AgtR1 occurred within intron regions, exemplifying an upregulation of genetic transcription for this gene. Nicotine increased angiotensin signaling in vitro. CB reactivity to angiotensin was increased in perinatal nicotine-exposed offspring compared with control offspring. Furthermore, CB denervation reduced arterial pressure because of suppressed efferent sympathetic activity in perinatal nicotine-exposed offspring. CONCLUSIONS Our data demonstrate that perinatal nicotine exposure adversely affects CB afferent sensing, which augments efferent sympathetic activity to increase vasoconstrictor signaling and induce hypertension. Targeting angiotensin signaling in the carotid bodies may provide a way to alleviate hypertension acquired by adverse maternal uterine environments in general and perinatal nicotine exposure in particular.
Collapse
Affiliation(s)
- Fengli Zhu
- The Lundquist Institute for Biomedical Innovation (F.Z., Z.W., K.D., H.M., J.Z., J.L., W.Y., V.K.R., N.J.), Harbor-UCLA Medical Center, Torrance
| | - Zhuqing Wang
- The Lundquist Institute for Biomedical Innovation (F.Z., Z.W., K.D., H.M., J.Z., J.L., W.Y., V.K.R., N.J.), Harbor-UCLA Medical Center, Torrance
| | - Kayla Davis
- The Lundquist Institute for Biomedical Innovation (F.Z., Z.W., K.D., H.M., J.Z., J.L., W.Y., V.K.R., N.J.), Harbor-UCLA Medical Center, Torrance
| | - Hayden McSwiggin
- The Lundquist Institute for Biomedical Innovation (F.Z., Z.W., K.D., H.M., J.Z., J.L., W.Y., V.K.R., N.J.), Harbor-UCLA Medical Center, Torrance
| | - Jekaterina Zyuzin
- The Lundquist Institute for Biomedical Innovation (F.Z., Z.W., K.D., H.M., J.Z., J.L., W.Y., V.K.R., N.J.), Harbor-UCLA Medical Center, Torrance
| | - Jie Liu
- The Lundquist Institute for Biomedical Innovation (F.Z., Z.W., K.D., H.M., J.Z., J.L., W.Y., V.K.R., N.J.), Harbor-UCLA Medical Center, Torrance
| | - Wei Yan
- The Lundquist Institute for Biomedical Innovation (F.Z., Z.W., K.D., H.M., J.Z., J.L., W.Y., V.K.R., N.J.), Harbor-UCLA Medical Center, Torrance
- Division of Metabolic Diseases and Translational Genomics (W.Y.), Harbor-UCLA Medical Center, Torrance
- David Geffen School of Medicine, University of California, Los Angeles (W.Y., V.K.R., N.J.)
| | - Virender K. Rehan
- The Lundquist Institute for Biomedical Innovation (F.Z., Z.W., K.D., H.M., J.Z., J.L., W.Y., V.K.R., N.J.), Harbor-UCLA Medical Center, Torrance
- Division of Neonatology (V.K.R.), Harbor-UCLA Medical Center, Torrance
- David Geffen School of Medicine, University of California, Los Angeles (W.Y., V.K.R., N.J.)
| | - Nicholas Jendzjowsky
- The Lundquist Institute for Biomedical Innovation (F.Z., Z.W., K.D., H.M., J.Z., J.L., W.Y., V.K.R., N.J.), Harbor-UCLA Medical Center, Torrance
- Division of Respiratory and Critical Care Medicine and Physiology (N.J.), Harbor-UCLA Medical Center, Torrance
- David Geffen School of Medicine, University of California, Los Angeles (W.Y., V.K.R., N.J.)
| |
Collapse
|
8
|
Chappell MC, Schaich CL, Busse LW, Clark Files D, Martin GS, Sevransky JE, Hinson JS, Rothman RE, Khanna AK. Higher circulating ACE2 and DPP3 but reduced ACE and angiotensinogen in hyperreninemic sepsis patients. Clin Sci (Lond) 2025; 139:43-53. [PMID: 39699964 DOI: 10.1042/cs20242168] [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: 10/11/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 12/21/2024]
Abstract
Sepsis and septic shock are global healthcare problems associated with high mortality rates. Activation of the renin-angiotensin-aldosterone system (RAAS) is an early event in sepsis, and elevated renin may be predictive of worse outcomes. In a subset of sepsis patients enrolled in the Vitamin C, Thiamine and Steroids in Sepsis (VICTAS) trial, elevated levels of active renin (median value > 189 pg/mL or 5.1 pM) at baseline (day 0) were strongly associated with mortality; however, corresponding plasma levels of the vasopressor hormone Angiotensin II were not substantially increased nor was Angiotensin II associated with disease severity. The current study assessed RAAS components that may impact the Angiotensin II response in control subjects, normal renin sepsis (NRS, renin < 5.1 pM) and high renin sepsis (HRS, renin > 5.1 pM) patients. NRS and HRS subjects exhibited a similar reduction in ACE (40%), but increased levels of ACE2 and DPP3. The ACE to DPP3 ratio was higher in controls but this relationship was reversed in both NRS and HRS subjects. Intact angiotensinogen was 50% lower in the HRS than control or NRS subjects, whereas the intact angiotensinogen to renin ratio was <10% of control or NRS subjects. We conclude that altered expression of ACE, ACE2, DPP3 and angiotensinogen may attenuate the expected increase in Angiotensin II, particularly in sepsis subjects with high renin concentrations.
Collapse
Affiliation(s)
- Mark C Chappell
- Hypertension Center, Wake Forest University School of Medicine, Winston-Salem, NC
| | | | | | - D Clark Files
- Department of Internal Medicine, Section of Pulmonary, Critical Care, Allergy, and Immunologic Diseases, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Greg S Martin
- Pulmonary and Critical Care, Emory University School of Medicine, Atlanta, GA
| | | | | | | | - Ashish K Khanna
- Hypertension Center, Wake Forest University School of Medicine, Winston-Salem, NC
- Department of Anesthesiology, Section on Critical Care Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
- Outcomes Research Consortium, Cleveland, OH
| |
Collapse
|
9
|
Westwood BM, Chappell MC. Angiotensin Converting Enzyme Hub Model of Peptide Flux Relating Calculated AT1 Receptor and Aldosterone Levels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608830. [PMID: 39713451 PMCID: PMC11661088 DOI: 10.1101/2024.08.20.608830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Utilizing data from the Vitamin C, Thiamine, and Steroids in Sepsis (VICTAS) Trial, this hub model was developed to limit seventeen Renin-Angiotensin-Aldosterone System (RAAS) components as three entrance and four exits, to facilitate the calculation of a model as one egress unknown, the angiotensin type 1 (AT1) receptor. Following previous evidence relating renin levels to mortality, this study found controls were more like sepsis patients with levels < renin quartile 1 (Q1) for calculated AT1, while more like sepsis patients with renin levels > quartile 3 (Q4) for measured aldosterone levels. Additionally differential discrete correlate summation (DCS) analysis indicates AT1, aldosterone and renin as major hub nodes in this independent DCS model of metabolic data inputs.
Collapse
|
10
|
Dominici FP, Gironacci MM, Narvaez Pardo JA. Therapeutic opportunities in targeting the protective arm of the renin-angiotensin system to improve insulin sensitivity: a mechanistic review. Hypertens Res 2024; 47:3397-3408. [PMID: 39363004 DOI: 10.1038/s41440-024-01909-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: 05/13/2024] [Revised: 08/04/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024]
Abstract
In recent years, the knowledge of the physiological and pathophysiological roles of the renin-angiotensin system (RAS) in glucose metabolism has advanced significantly. It is now well-established that blockade of the angiotensin AT1 receptor (AT1R) improves insulin sensitivity. Activation of the AT2 receptor (AT2R) and the MAS receptor are significant contributors to this beneficial effect. Elevated availability of angiotensin (Ang) II) for interaction with the AT2R and increased Ang-(1-7) formation during AT1R blockade mediate these effects. The ongoing development of selective AT2R agonists, such as compound 21 and the novel Ang III peptidomimetics, has significantly advanced the exploration of the role of AT2R in metabolism and its potential as a therapeutic target. These agents show promise, particularly when RAS inhibition is contraindicated. Additionally, other RAS peptides, including Ang IV, des-Asp-Ang I, Ang-(1-9), and alamandine, hold therapeutic capability for addressing metabolic disturbances linked to type 2 diabetes. The possibility of AT2R heteromerization with either AT1R or MAS receptor offers an exciting area for future research, particularly concerning therapeutic strategies to improve glycemic control. This review focuses on therapeutic opportunities to improve insulin sensitivity, taking advantage of the protective arm of the RAS.
Collapse
Affiliation(s)
- Fernando P Dominici
- Departamento de Química Biológica and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Mariela M Gironacci
- Departamento de Química Biológica and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jorge A Narvaez Pardo
- Departamento de Química Biológica and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
11
|
Schofield LG, Endacott SK, Delforce SJ, Lumbers ER, Pringle KG. Importance of the (Pro)renin Receptor in Activating the Renin-Angiotensin System During Normotensive and Preeclamptic Pregnancies. Curr Hypertens Rep 2024; 26:483-495. [PMID: 39093387 PMCID: PMC11455731 DOI: 10.1007/s11906-024-01316-1] [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] [Accepted: 07/02/2024] [Indexed: 08/04/2024]
Abstract
PURPOSE OF REVIEW For a healthy pregnancy to occur, a controlled interplay between the maternal circulating renin-angiotensin-aldosterone system (RAAS), placental renin-angiotensin system (RAS) and intrarenal renin-angiotensin system (iRAS) is necessary. Functionally, both the RAAS and iRAS interact to maintain blood pressure and cardiac output, as well as fluid and electrolyte balance. The placental RAS is important for placental development while also influencing the maternal circulating RAAS and iRAS. This narrative review concentrates on the (pro)renin receptor ((P)RR) and its soluble form (s(P)RR) in the context of the hypertensive pregnancy pathology, preeclampsia. RECENT FINDINGS The (P)RR and the s(P)RR have become of particular interest as not only can they activate prorenin and renin, thus influencing levels of angiotensin II (Ang II), but s(P)RR has now been shown to directly interact with and stimulate the Angiotensin II type 1 receptor (AT1R). Levels of both placental (P)RR and maternal circulating s(P)RR are elevated in patients with preeclampsia. Furthermore, s(P)RR has been shown to increase blood pressure in non-pregnant and pregnant rats and mice. In preeclamptic pregnancies, which are characterised by maternal hypertension and impaired placental development and function, we propose that there is enhanced secretion of s(P)RR from the placenta into the maternal circulation. Due to its ability to both activate prorenin and act as an AT1R agonist, excess maternal circulating s(P)RR can act on both the maternal vasculature, and the kidney, leading to RAS over-activation. This results in dysregulation of the maternal circulating RAAS and overactivation of the iRAS, contributing to maternal hypertension, renal damage, and secondary changes to neurohumoral regulation of fluid and electrolyte balance, ultimately contributing to the pathophysiology of preeclampsia.
Collapse
Affiliation(s)
- Lachlan G Schofield
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, N.S.W, 2308, Australia
- Womens Health Research Program, Hunter Medical Research Institute, New Lambton Heights, N.S.W, 2305, Australia
- Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton, N.S.W, 2305, Australia
| | - Saije K Endacott
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, N.S.W, 2308, Australia
- Womens Health Research Program, Hunter Medical Research Institute, New Lambton Heights, N.S.W, 2305, Australia
- Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton, N.S.W, 2305, Australia
| | - Sarah J Delforce
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, N.S.W, 2308, Australia
- Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton, N.S.W, 2305, Australia
| | - Eugenie R Lumbers
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, N.S.W, 2308, Australia
- Womens Health Research Program, Hunter Medical Research Institute, New Lambton Heights, N.S.W, 2305, Australia
- Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton, N.S.W, 2305, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, N.S.W, 2308, Australia.
- Womens Health Research Program, Hunter Medical Research Institute, New Lambton Heights, N.S.W, 2305, Australia.
- Hunter Medical Research Institute, Lot 1 Kookaburra Circuit, New Lambton, N.S.W, 2305, Australia.
| |
Collapse
|
12
|
Dos Santos L, Favaroni Mendes Salgado Ribeiro LA, Febba Gomes AC, Azinheira Nobrega Cruz N, Gonçalves de Oliveira LC, Cenedeze MA, Tedesco Silva Junior H, Medina Pestana JO, Casarini DE. ACE and ACE2 activities and polymorphisms assessment: A populational study from Ipaussu (SP, Brazil) during the COVID-19 pandemic. Life Sci 2024; 358:123157. [PMID: 39437850 DOI: 10.1016/j.lfs.2024.123157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
AIM The angiotensin-converting enzyme 2 (ACE2) and its homolog, the angiotensin converting enzyme 1 (ACE), are involved in COVID-19 physiopathology. Alterations in the enzymatic structure, expression, and/or activity may influence the risk of infection and severity of disease. For this reason, we aimed to identify different allelic forms of ACE2 G8790A and ACE I/D polymorphisms in a Brazilian cohort and evaluate their impact on ACE and ACE2 activities and their association with COVID-19 susceptibility and severity. MAIN METHODS A total of 549 COVID-19-negative and 270 COVID-19-positive participants from Ipaussu, Sao Paulo, Brazil, were recruited. ACE2 and ACE activities were measured by fluorogenic assays using MCA-Ala-Pro-Lys(Dnp) as the substrate for ACE2 and Z-Phe-His-Leu-OH (Z-FHL) and Hippuryl-His-Leu-OH (h-HL) as substrates for ACE. Genomic DNA was extracted from EDTA-peripheral blood, and the regions of the genes containing ACE2 G8790A and ACE I/D polymorphisms were amplified by PCR-restriction fragment length polymorphism and real-time PCR, respectively. KEY FINDINGS The G allele of ACE2 G8790A polymorphism and D allele of ACE I/D polymorphism are associated with increased ACE and ACE2 activities. ACE activity ratio (Z-FHL/h-HL), an inflammatory marker, is increased in women with GG genotype and COVID-19-positive diagnosis. SIGNIFICANCE For the first time, it was demonstrated that in females, the GG genotype is associated with increased ACE activity ratio (Z-FHL/h-HL) in the COVID-19-positive group. Elevated ACE activity ratio (Z-FHL/h-HL) is highly linked to inflammation and may justify the associations between the G genotype and COVID-19 severity of symptoms and outcomes.
Collapse
Affiliation(s)
- Lilian Dos Santos
- Department of Medicine, Discipline of Nephrology, University Federal of Sao Paulo, Sao Paulo, Brazil
| | | | | | | | | | - Marcos Antonio Cenedeze
- Department of Medicine, Discipline of Nephrology, University Federal of Sao Paulo, Sao Paulo, Brazil
| | | | - José Osmar Medina Pestana
- Department of Medicine, Discipline of Nephrology, University Federal of Sao Paulo, Sao Paulo, Brazil
| | - Dulce Elena Casarini
- Department of Medicine, Discipline of Nephrology, University Federal of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
13
|
Islam MA, Ford Versypt AN. Mathematical Modeling of Impacts of Patient Differences on Renin-Angiotensin System and Applications to COVID-19 Lung Fibrosis Outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2022.11.06.515367. [PMID: 36380760 PMCID: PMC9665336 DOI: 10.1101/2022.11.06.515367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Patient-specific premorbidity, age, and sex are significant heterogeneous factors that influence the severe manifestation of lung diseases, including COVID-19 fibrosis. The renin-angiotensin system (RAS) plays a prominent role in regulating the effects of these factors. Recent evidence shows patient-specific alterations of RAS homeostasis concentrations with premorbidity and the expression level of angiotensin-converting enzyme 2 (ACE2) during COVID-19. However, conflicting evidence suggests decreases, increases, or no changes in RAS peptides after SARS-CoV-2 infection. In addition, detailed mechanisms connecting the patient-specific conditions before infection to infection-induced RAS alterations are still unknown. Here, a multiscale computational model was developed to quantify the systemic contribution of heterogeneous factors of RAS during COVID-19. Three submodels were connected-an agent-based model for in-host COVID-19 response in the lung tissue, a RAS dynamics model, and a fibrosis dynamics model to investigate the effects of patient-group-specific factors in the systemic alteration of RAS and collagen deposition in the lung. The model results indicated cell death due to inflammatory response as a major contributor to the reduction of ACE and ACE2. In contrast, there were no significant changes in ACE2 dynamics due to viral-bound internalization of ACE2. The model explained possible mechanisms for conflicting evidence of patient-group-specific changes in RAS peptides in previously published studies. Simulated results were consistent with reported RAS peptide values for SARS-CoV-2-negative and SARS-CoV-2-positive patients. RAS peptides decreased for all virtual patient groups with aging in both sexes. In contrast, large variations in the magnitude of reduction were observed between male and female virtual patients in the older and middle-aged groups. The patient-specific variations in homeostasis RAS peptide concentrations of SARS-CoV-2-negative patients also affected the dynamics of RAS during infection. The model results also showed that feedback between RAS signaling and renin dynamics could restore ANGI homeostasis concentration but failed to restore homeostasis values of RAS peptides downstream of ANGI. In addition, the results showed that ACE2 variations with age and sex significantly altered the concentrations of RAS peptides and led to collagen deposition with slight variations depending on age and sex. This model may find further applications in patient-specific calibrations of tissue models for acute and chronic lung diseases to develop personalized treatments.
Collapse
|
14
|
Michaëlsson K, Lemming EW, Larsson SC, Höijer J, Melhus H, Svennblad B, Baron JA, Wolk A, Byberg L. Non-fermented and fermented milk intake in relation to risk of ischemic heart disease and to circulating cardiometabolic proteins in swedish women and men: Two prospective longitudinal cohort studies with 100,775 participants. BMC Med 2024; 22:483. [PMID: 39511582 PMCID: PMC11546556 DOI: 10.1186/s12916-024-03651-1] [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: 04/20/2024] [Accepted: 09/24/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND The effect of milk on the risk of ischemic heart disease (IHD) and acute myocardial infarction (MI) is unclear. We aimed to examine the association between non-fermented and fermented milk consumption on these endpoints and investigate the relationship between milk intake and cardiometabolic-related proteins in plasma. METHODS Our study is based on two Swedish prospective cohort studies that included 59,998 women and 40,777 men without IHD or cancer at baseline who provided repeated measures of diet and lifestyle factors and plasma proteomics data in two subcohorts. Through registry linkage, 17,896 cases with IHD were documented during up to 33 years of follow-up, including 10,714 with MI. We used time-updated multivariable Cox regression analysis to examine non-fermented or fermented milk intake with time to IHD or MI. Using high-throughput multiplex immunoassays, 276 cardiometabolic plasma proteins were measured in two subcohorts. We applied multivariable-adjusted regression models using a discovery-replication design to examine protein associations with increasing consumption of non-fermented or fermented milk. RESULTS The results for non-fermented milk differed by sex (p-value for interaction = 0.01). In women, we found a pattern of successively greater risk of IHD and MI at non-fermented milk intake levels higher than 1.5 glasses/day. Compared with an intake of 0.5 glass/day (100 mL/day), non-fermented milk intake of 2 glasses/day in women conferred a multivariable-adjusted hazard ratio (HR) of 1.05 (95% CI 1.01-1.08) for IHD, an intake of 3 glasses/day an HR of 1.12 (95% CI 1.06-1.19), and an intake of 4 glasses/day an HR of 1.21 (95% CI 1.10-1.32). Findings were similar for whole, medium-fat, and low-fat milk. We did not detect higher risks of IHD with increasing milk intakes in men. Fermented milk intake was unrelated to the risk of IHD or MI in either sex. Increasing non-fermented milk intake in women was robustly associated with a higher concentration of plasma ACE2 and a lower concentration of FGF21. CONCLUSIONS We show a positive association between high amounts of non-fermented milk intake and IHD in women but not men. We suggest metabolic pathways related to ACE2 and FGF21 potentially underlie the association.
Collapse
Affiliation(s)
- Karl Michaëlsson
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| | - Eva Warensjö Lemming
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Susanna C Larsson
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Höijer
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Håkan Melhus
- Clinical Pharmacology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Bodil Svennblad
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - John A Baron
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Alicja Wolk
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Liisa Byberg
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
15
|
Chappell MC, Schaich CL, Busse LW, Martin GS, Sevransky JE, Hinson JK, Khanna AK. Stronger association of intact angiotensinogen with mortality than lactate or renin in critical illness: post-hoc analysis from the VICTAS trial. Crit Care 2024; 28:333. [PMID: 39402593 PMCID: PMC11472595 DOI: 10.1186/s13054-024-05120-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Sepsis and septic shock remain global healthcare problems associated with high mortality rates despite best therapy efforts. Circulating biomarkers may identify those patients at risk for poor outcomes, however, current biomarkers, most prominently lactate, are non-specific and have an inconsistent impact on prognosis and/or disease management. Activation of the renin-angiotensin- system (RAS) is an early event in sepsis patients and elevated levels of circulating renin are more predictive of worse outcomes than lactate. The precursor protein Angiotensinogen is another key component of the circulating RAS; it is the only known substrate for renin and the ultimate source of the vasopressor Angiotensin II (Ang II). We postulate that lower Angiotensinogen concentrations may reflect a dysfunctional RAS characterized by high renin concentrations but attenuated Ang II generation, which is disproportionate to the high renin response and may compromise adequate support of blood pressure and tissue perfusion in septic patients. The current study compared the association between serum Angiotensinogen with mortality to that of lactate and renin in the VICTAS cohort of sepsis patients at baseline (day 0) by receiver operating characteristic (ROC) and Kaplan-Meier curve analyses. Serum concentration of Angiotensinogen was more strongly associated with 30-day mortality than either the serum concentrations of renin or lactate in sepsis patients. Moreover, the clinical assessment of Angiotensinogen may have distinct advantages over the typical measures of renin. The assessment of intact Angiotensinogen may potentially facilitate more precise therapeutic approaches (including exogenous angiotensin II) to restore a dysfunctional RAS and improve patient outcomes. Additional prospective validation studies are clearly required for this biomarker in the future.
Collapse
Affiliation(s)
- Mark C Chappell
- Hypertension Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Christopher L Schaich
- Hypertension Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Laurence W Busse
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Greg S Martin
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathan E Sevransky
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Ashish K Khanna
- Hypertension Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
- Department of Anesthesiology, Section On Critical Care Medicine, Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157-1009, USA.
- Outcomes Research Consortium, Houston, TX, USA.
| |
Collapse
|
16
|
Chen D, Zeng S, Liu Q. Changes in nonfunctional adrenal incidentaloma after COVID-19 infection and a model for predicting benign and malignant adrenal incidentaloma. Front Endocrinol (Lausanne) 2024; 15:1374282. [PMID: 39286271 PMCID: PMC11402735 DOI: 10.3389/fendo.2024.1374282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 07/24/2024] [Indexed: 09/19/2024] Open
Abstract
Aims To compare nonfunctional adrenal incidentalomas (NFAI) in individuals with and without a history of COVID-19 infection, while also establishing predictive models for distinguishing between benign and malignant adrenal incidentalomas (AI). Methods A retrospective collection of data from patients with AI who underwent surgery and were verified in our hospital between April 2022 and June 2023 was conducted. A total of 121 patients were included in the study. Demographic information, tumor characteristics, functional indicators, and complications were compared among the patients. Statistical analyses utilized the t-test for continuous variables and Pearson chi-square test or Fisher's exact test for categorical variables. Results Patients with COVID-19 exhibited a higher prevalence of obesity (84.2% vs. 63.3%, P=0.048) and elevated direct bilirubin (DBIL) levels (44.1% vs. 19.2%, P=0.043) compared to those without COVID-19. Moreover, patients with Malignant AI, in contrast to Benign AI, showed higher normal total protein (TP) levels (28.8% vs. 57.1%, P=0.016) and larger tumor sizes (20 vs. 32.5mm, P=0.009). Univariate analysis identified low TP (OR=0.303, 95% CI=0.111-0.825, P=0.020) and tumor size (OR=1.045, 95% CI=1.011-1.080, P=0.009) as potential risk factors for multivariate analysis. A predictive model comprising clinical risk factors (tumor size and low TP) demonstrated an AUC of 0.754 (95% CI, 0.603-0.904) with a sensitivity of 0.75 and specificity of 0.775. The calibration curve revealed a bias-corrected AUC of 0.77. Conclusion No discernible differences in the clinical manifestations of adrenal incidentalomas were observed between cases with and without a history of COVID-19 infection. However, AI with larger tumor diameters and lower than normal levels of total protein exhibited a more pronounced malignant potential.
Collapse
Affiliation(s)
- Danlei Chen
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
- Department of Urology, First People's Hospital of Yunnan Province, Kunming, China
| | - Sheng Zeng
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
17
|
Fernando RJ, Royster RL, Ferrario CM, Saha AK, Ahmad S, Henshaw DS, Kittner SL, Talbott AL, Khanna AK, Morris BN, Groban L, Templeton TW. Angiotensin II treatment of hypotension in noncardiac surgery: an initial dose-finding study. Br J Anaesth 2024; 133:667-670. [PMID: 38908951 DOI: 10.1016/j.bja.2024.04.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/14/2024] [Accepted: 04/28/2024] [Indexed: 06/24/2024] Open
Affiliation(s)
- Rohesh J Fernando
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | - Roger L Royster
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Carlos M Ferrario
- Department of General Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Amit K Saha
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Sarfaraz Ahmad
- Department of General Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Daryl S Henshaw
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Sarah L Kittner
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Ashley L Talbott
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Ashish K Khanna
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Perioperative Outcomes and Informatics Collaborative, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Outcomes Research Consortium, Cleveland, OH, USA
| | - Benjamin N Morris
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Thomas W Templeton
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
18
|
Wopperer FJ, Olinger E, Wiesener A, Broeker KAE, Knaup KX, Schaefer JT, Galiano M, Schneider K, Schiffer M, Büttner-Herold M, Reis A, Schmieder R, Pasutto F, Hilgers KF, Poglitsch M, Ziegler C, Shoemaker R, Sayer JA, Wiesener MS. Progressive Kidney Failure by Angiotensinogen Inactivation in the Germline. Hypertension 2024; 81:1857-1868. [PMID: 39005223 DOI: 10.1161/hypertensionaha.124.22806] [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: 02/05/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Autosomal recessive renal tubular dysgenesis is a rare, usually fatal inherited disorder of the renin-angiotensis system (RAS). Herein, we report an adolescent individual experiencing an unknown chronic kidney disease and aim to provide novel insights into disease mechanisms. METHODS Exome sequencing for a gene panel associated with renal disease was performed. The RAS was assessed by comprehensive biochemical analysis in blood. Renin expression was determined in primary tubular cells by quantitative polymerase chain reaction and in situ hybridization on kidney biopsy samples. Allele frequencies of heterozygous and biallelic deleterious variants were determined by analysis of the Genomics England 100,000 Genomes Project. RESULTS The patient was delivered prematurely after oligohydramnios was detected during pregnancy. Postnatally, he recovered from third-degree acute kidney injury but developed chronic kidney disease stage G3b over time. Exome sequencing revealed a previously reported pathogenic homozygous missense variant, p.(Arg375Gln), in the AGT (angiotensinogen) gene. Blood AGT concentrations were low, but plasma renin concentration and gene expression in kidney biopsy, vascular, and tubular cells revealed strong upregulation of renin. Angiotensin II and aldosterone in blood were not abnormally elevated. CONCLUSIONS Renal tubular dysgenesis may present as chronic kidney disease with a variable phenotype, necessitating broad genetic analysis for diagnosis. Functional analysis of the RAS in a patient with AGT mutation revealed novel insights regarding compensatory upregulation of renin in vascular and tubular cells of the kidney and in plasma in response to depletion of AGT substrate as a source of Ang II (similarly observed with hepatic AGT silencing for the treatment of hypertension).
Collapse
Affiliation(s)
- Florian J Wopperer
- Department of Nephrology and Hypertension (F.J.W., K.X.K., K.S., M.S., R. Schmieder, K.F.H., M.S.W.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Eric Olinger
- Center for Human Genetics, Cliniques universitaires Saint-Luc, Brussels, Belgium (E.O.)
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom (E.O., J.A.S.)
| | - Antje Wiesener
- Institute of Human Genetics (A.W., A.R., F.P.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | | | - Karl X Knaup
- Department of Nephrology and Hypertension (F.J.W., K.X.K., K.S., M.S., R. Schmieder, K.F.H., M.S.W.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Jan T Schaefer
- Department of Pediatrics and Adolescent Medicine (J.T.S., M.G.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Matthias Galiano
- Department of Pediatrics and Adolescent Medicine (J.T.S., M.G.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Karen Schneider
- Department of Nephrology and Hypertension (F.J.W., K.X.K., K.S., M.S., R. Schmieder, K.F.H., M.S.W.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension (F.J.W., K.X.K., K.S., M.S., R. Schmieder, K.F.H., M.S.W.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Maike Büttner-Herold
- Department of Nephropathology, Institute of Pathology (M.B.-H.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - André Reis
- Institute of Human Genetics (A.W., A.R., F.P.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Roland Schmieder
- Department of Nephrology and Hypertension (F.J.W., K.X.K., K.S., M.S., R. Schmieder, K.F.H., M.S.W.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Francesca Pasutto
- Institute of Human Genetics (A.W., A.R., F.P.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Karl F Hilgers
- Department of Nephrology and Hypertension (F.J.W., K.X.K., K.S., M.S., R. Schmieder, K.F.H., M.S.W.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | | | | | - Robin Shoemaker
- Department of Pediatrics, University of Kentucky, Lexington (R. Shoemaker)
| | - John A Sayer
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom (E.O., J.A.S.)
| | - Michael S Wiesener
- Department of Nephrology and Hypertension (F.J.W., K.X.K., K.S., M.S., R. Schmieder, K.F.H., M.S.W.), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| |
Collapse
|
19
|
Maurer J, de Groot A, Martin L, Grouzmann E, Wuerzner G, Eugster PJ. Quantification of endogenous Angiotensin 1-10, 1-9, 1-8, 1-7, and 1-5 in human plasma using micro-UHPLC-MS/MS: Outlining the importance of the pre-analytics for reliable results. J Pharm Biomed Anal 2024; 243:116101. [PMID: 38489957 DOI: 10.1016/j.jpba.2024.116101] [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: 01/12/2024] [Revised: 02/26/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024]
Abstract
Angiotensin peptides (ANGs) play a central role in the renin-angiotensin-aldosterone system, rendering them interesting biomarkers associated with hypertension. Precise quantification of circulating ANGs holds the potential to assess the activity of angiotensin-converting enzyme (ACE), a key protease targeted by widely prescribed drugs, namely ACE inhibitors. This ability could pave the way for personalised medicine, offering insights into the prescription of inhibitors targeting either the proteases or the receptors within the system. Despite recent developments in liquid chromatography-mass spectrometry (LC-MS) methods for measuring circulating ANG concentrations, comprehensive stability studies of ANGs in human plasma are absent in the literature, raising concerns about the reliability of measured concentrations and their link to clinical conditions. To address this critical gap, we conducted an exhaustive evaluation of the pre-analytical stability of ANG1-10, ANG1-9, ANG1-8, ANG1-7, and ANG1-5. By employing surfactants to mitigate non-specific adsorption and a dedicated mix of protease inhibitors to limit protease activity, we established an MS-based assay for these five peptides. We used this method to quantify circulating concentrations of ANGs in the plasma of 11 healthy donors and 3 patients under kidney dialysis. Our findings revealed that ANG1-10 and ANG1-8 circulate at concentrations ranging from 1 to 10 pM in healthy subjects and exhibit a high degree of correlation. Notably, ANG1-9, ANG1-7, and ANG1-5 were undetectable in any of the 14 patients, despite a sub-picomolar limit of detection. This strikingly contrasts with the reference concentrations reported in the literature, which typically fall within the picomolar range. In light of these discrepancies, we strongly advocate for rigorous pre-analytical considerations and comprehensive stability studies to ensure reliable results. We emphasise the pivotal role of heightened pre-analytical awareness within the clinical chemistry community, and we hope for continued growth in this critical area.
Collapse
Affiliation(s)
- Jonathan Maurer
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Anke de Groot
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Léon Martin
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Eric Grouzmann
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Grégoire Wuerzner
- Service of Hypertension and Nephrology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Philippe J Eugster
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
20
|
Picod A, Garcia B, Van Lier D, Pickkers P, Herpain A, Mebazaa A, Azibani F. Impaired angiotensin II signaling in septic shock. Ann Intensive Care 2024; 14:89. [PMID: 38877367 PMCID: PMC11178728 DOI: 10.1186/s13613-024-01325-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/29/2024] [Indexed: 06/16/2024] Open
Abstract
Recent years have seen a resurgence of interest for the renin-angiotensin-aldosterone system in critically ill patients. Emerging data suggest that this vital homeostatic system, which plays a crucial role in maintaining systemic and renal hemodynamics during stressful conditions, is altered in septic shock, ultimately leading to impaired angiotensin II-angiotensin II type 1 receptor signaling. Indeed, available evidence from both experimental models and human studies indicates that alterations in the renin-angiotensin-aldosterone system during septic shock can occur at three distinct levels: 1. Impaired generation of angiotensin II, possibly attributable to defects in angiotensin-converting enzyme activity; 2. Enhanced degradation of angiotensin II by peptidases; and/or 3. Unavailability of angiotensin II type 1 receptor due to internalization or reduced synthesis. These alterations can occur either independently or in combination, ultimately leading to an uncoupling between the renin-angiotensin-aldosterone system input and downstream angiotensin II type 1 receptor signaling. It remains unclear whether exogenous angiotensin II infusion can adequately address all these mechanisms, and additional interventions may be required. These observations open a new avenue of research and offer the potential for novel therapeutic strategies to improve patient prognosis. In the near future, a deeper understanding of renin-angiotensin-aldosterone system alterations in septic shock should help to decipher patients' phenotypes and to implement targeted interventions.
Collapse
Affiliation(s)
- Adrien Picod
- INSERM, UMR-S 942 MASCOT-Université Paris-Cité, Paris, France.
| | - Bruno Garcia
- Department of Intensive Care Medicine, Centre Hospitalier Universitaire de Lille, Lille, France
- Experimental Laboratory of Intensive Care, Université Libre de Bruxelles, Brussels, Belgium
| | - Dirk Van Lier
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Antoine Herpain
- Experimental Laboratory of Intensive Care, Université Libre de Bruxelles, Brussels, Belgium
- Department of Intensive Care Medicine, St. Pierre University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Alexandre Mebazaa
- INSERM, UMR-S 942 MASCOT-Université Paris-Cité, Paris, France
- Department of Anesthesiology, Burns and Critical Care, Hopitaux Saint-Louis-Lariboisière, AP-HP, Paris, France
| | - Feriel Azibani
- INSERM, UMR-S 942 MASCOT-Université Paris-Cité, Paris, France
| |
Collapse
|
21
|
Schofield LG, Delforce SJ, Pryor JC, Endacott SK, Lumbers ER, Marshall SA, Pringle KG. The soluble (pro)renin receptor promotes a preeclampsia-like phenotype both in vitro and in vivo. Hypertens Res 2024; 47:1627-1641. [PMID: 38605139 PMCID: PMC11150152 DOI: 10.1038/s41440-024-01678-8] [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: 11/20/2023] [Revised: 03/14/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024]
Abstract
Preeclampsia is classified as new-onset hypertension coupled with gross endothelial dysfunction. Placental (pro)renin receptor ((P)RR) and plasma soluble (P)RR (s(P)RR) are elevated in patients with preeclampsia. Thus, we aimed to interrogate the role (P)RR may play in the pathogenesis of preeclampsia. Human uterine microvascular endothelial cells (HUtMECs, n = 4) were cultured with either; vehicle (PBS), 25-100 nM recombinant s(P)RR, or 10 ng/ml TNF-a (positive control) for 24 h. Conditioned media and cells were assessed for endothelial dysfunction markers via qPCR, ELISA, and immunoblot. Angiogenic capacity was assessed through tube formation and adhesion assays. Additionally, pregnant rats were injected with an adenovirus overexpressing s(P)RR from mid-pregnancy (day 8.5), until term (n = 6-7 dams/treatment). Maternal and fetal tissues were assessed. HUtMECs treated with recombinant s(P)RR displayed increased expression of endothelial dysfunction makers including vascular cell adhesion molecule-1, intracellular adhesion molecule-1, and endothelin-1 mRNA expression (P = 0.003, P = 0.001, P = 0.009, respectively), along with elevated endothelin-1 protein secretion (P < 0.001) compared with controls. Recombinant s(P)RR impaired angiogenic capacity decreasing the number of branches, total branch length, and mesh area (P < 0.001, P = 0.004, and P = 0.009, respectively), while also increasing vascular adhesion (P = 0.032). +ADV rats exhibited increased systolic (P = 0.001), diastolic (P = 0.010), and mean arterial pressures (P = 0.012), compared with -ADV pregnancies. Renal arteries from +ADV-treated rats had decreased sensitivity to acetylcholine-induced relaxation (P = 0.030), compared with -ADV pregnancies. Our data show that treatment with s(P)RR caused hypertension and growth restriction in vivo and caused marked endothelial dysfunction in vitro. These findings demonstrate the significant adverse actions of s(P)RR on vascular dysfunction that is characteristic of the preeclamptic phenotype.
Collapse
Affiliation(s)
- Lachlan G Schofield
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Sarah J Delforce
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Jennifer C Pryor
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- National Health & Medical Research Council (NHMRC) Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
| | - Saije K Endacott
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Eugenie R Lumbers
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Sarah A Marshall
- Department of Obstetrics and Gynaecology, The Ritchie Centre, School of Clinical Sciences, Monash University and The Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia.
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.
| |
Collapse
|
22
|
de Cavanagh EMV, Inserra F, Ferder L. Renin-angiotensin system inhibitors positively impact on multiple aging regulatory pathways: Could they be used to protect against human aging? Physiol Rep 2024; 12:e16094. [PMID: 38924381 PMCID: PMC11200104 DOI: 10.14814/phy2.16094] [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: 02/22/2024] [Revised: 05/18/2024] [Accepted: 05/18/2024] [Indexed: 06/28/2024] Open
Abstract
The renin-angiotensin system (RAS)-a classical blood pressure regulator-largely contributes to healthy organ development and function. Besides, RAS activation promotes age-related changes and age-associated diseases, which are attenuated/abolished by RAS-blockade in several mammalian species. RAS-blockers also increase rodent lifespan. In previous work, we discussed how RAS-blockade downregulates mTOR and growth hormone/IGF-1 signaling, and stimulates AMPK activity (together with klotho, sirtuin, and vitamin D-receptor upregulation), and proposed that at least some of RAS-blockade's aging benefits are mediated through regulation of these intermediaries and their signaling to mitochondria. Here, we included RAS-blockade's impact on other aging regulatory pathways, that is, TGF-ß, NF-kB, PI3K, MAPK, PKC, Notch, and Wnt, all of which affect mitochondria. No direct evidence is available on RAS/RAS-blockade-aging regulatory pathway-mitochondria interactions. However, existing results allow to conjecture that RAS-blockers neutralize mitochondrial dysfunction by acting on the discussed pathways. The reviewed evidence led us to propose that the foundation is laid for conducting clinical trials aimed at testing whether angiotensin-converting enzyme inhibitors (ACEi) or angiotensin receptor blockers (ARB)-even at subclinical doses-offer the possibility to live longer and in better health. As ACEi and ARB are low cost and well-tolerated anti-hypertension therapies in use for over 35 years, investigating their administration to attenuate/prevent aging effects seems simple to implement.
Collapse
Affiliation(s)
| | - Felipe Inserra
- Department of MedicineMaimonides UniversityBuenos AiresArgentina
- Master of Vascular Mechanics and Arterial Hypertension, Postgraduate DepartmentAustral UniversityPilarArgentina
| | - León Ferder
- Department of MedicineMaimonides UniversityBuenos AiresArgentina
| |
Collapse
|
23
|
Schaich CL, Leisman DE, Goldberg MB, Filbin MR, Khanna AK, Chappell MC. Dysfunction of the renin-angiotensin-aldosterone system in human septic shock. Peptides 2024; 176:171201. [PMID: 38555976 PMCID: PMC11060897 DOI: 10.1016/j.peptides.2024.171201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
Sepsis and septic shock are global healthcare problems associated with mortality rates of up to 40% despite optimal standard-of-care therapy and constitute the primary cause of death in intensive care units worldwide. Circulating biomarkers of septic shock severity may represent a clinically relevant approach to individualize those patients at risk for worse outcomes early in the course of the disease, which may facilitate early and more precise interventions to improve the clinical course. However, currently used septic shock biomarkers, including lactate, may be non-specific and have variable impact on prognosis and/or disease management. Activation of the renin-angiotensin-aldosterone system (RAAS) is likely an early event in septic shock, and studies suggest that an elevated level of renin, the early and committed step in the RAAS cascade, is a better predictor of worse outcomes in septic shock, including mortality, than the current standard-of-care measure of lactate. Despite a robust increase in renin, other elements of the RAAS, including endogenous levels of Ang II, may fail to sufficiently increase to maintain blood pressure, tissue perfusion, and protective immune responses in septic shock patients. We review the current clinical literature regarding the dysfunction of the RAAS in septic shock and potential therapeutic approaches to improve clinical outcomes.
Collapse
Affiliation(s)
- Christopher L Schaich
- Hypertension & Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Daniel E Leisman
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Marcia B Goldberg
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Micheal R Filbin
- Department of Emergency Medicine, Massachusetts General Hospital,Boston, MA, USA
| | - Ashish K Khanna
- Hypertension & Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Department of Anesthesiology, Section on Critical Care Medicine, Atrium Health Wake Forest Baptist Medical Center, USA; Outcomes Research Consortium, Cleveland, OH, USA
| | - Mark C Chappell
- Hypertension & Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
24
|
Kotani Y, Chappell M, Landoni G, Zarbock A, Bellomo R, Khanna AK. Renin in critically ill patients. Ann Intensive Care 2024; 14:79. [PMID: 38775999 PMCID: PMC11111649 DOI: 10.1186/s13613-024-01304-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024] Open
Abstract
The renin-angiotensin system (RAS) constitutes one of the principal mechanisms to maintain hemodynamic and fluid homeostasis. However, most research until now on RAS primarily focuses on its relationship with hypertension and its role in critically ill hypotensive populations is not well understood. With the approval of angiotensin II (Ang II) in the United States and Europe, following a phase 3 randomized controlled trial showing efficacy in catecholamine-resistant vasodilatory shock, there is growing interest in RAS in critically ill patients. Among the fundamental components of RAS, renin acts as the initial stimulus for the entire system. In the context of hypotension, its release increases in response to low blood pressure sensed by renal baroreceptors and attenuated negative Ang II feedback loop. Thus, elevated renin could reflect disease severity and predict poor outcomes. Studies investigating this hypothesis have validated the prognostic accuracy of renin in various critically ill populations, with several reports indicating its superiority to lactate for mortality prediction. Accordingly, renin reduction has been used to assess the effectiveness of Ang II administration. Furthermore, renin holds potential to identify patients who might benefit from Ang II treatment, potentially paving the way for personalized vasopressor management. Despite these promising data, most available evidence is derived from retrospective analysis and necessitates prospective confirmation. The absence of a rapid, point-of-care and reliable renin assay presents another hurdle to its integration into routine clinical practice. This narrative review aims to describe the current understanding and future directions of renin as a biomarker during resuscitation of critically ill patients.
Collapse
Affiliation(s)
- Yuki Kotani
- Department of Intensive Care Medicine, Kameda Medical Center, Kamogawa, Japan
| | - Mark Chappell
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Muenster, Muenster, Germany
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Hospital, Melbourne, Australia
- Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, Australia
- Department of Critical Care, Melbourne Medical School, The University of Melbourne, Melbourne, Australia
| | - Ashish K Khanna
- Section On Critical Care Medicine, Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
- Perioperative Outcomes and Informatics Collaborative, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
- Outcomes Research Consortium, Cleveland, OH, 44195, USA.
| |
Collapse
|
25
|
Liu J, Song W, Gao X, Sun J, Liu C, Fang L, Wang J, Shi J, Leng Y, Liu X, Min W. A combined in vitro and in silico study of the inhibitory mechanism of angiotensin-converting enzyme with peanut peptides. Int J Biol Macromol 2024; 268:131901. [PMID: 38677685 DOI: 10.1016/j.ijbiomac.2024.131901] [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: 01/08/2024] [Revised: 04/06/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
Food-derived peptides with low molecular weight, high bioavailability, and good absorptivity have been exploited as angiotensin-converting enzyme (ACE) inhibitors. In the present study, in-vitro inhibition kinetics of peanut peptides, in silico screening, validation of ACE inhibitory activity, molecular dynamics (MD) simulations, and HUVEC cells were performed to systematically identify the inhibitory mechanism of ACE interacting with peanut peptides. The results indicate that FPHPP, FPHY, and FPHFD peptides have good thermal, pH, and digestive stability. MD trajectories elucidate the dynamic correlation between peptides and ACE and verify the specific binding interaction. Noteworthily, FPHPP is the best inhibitor with a strongest binding affinity and significantly increases NO, SOD production, and AT2R expression, and decreases ROS, MDA, ET-1 levels, ACE, and AT1R accumulation in Ang II-injury HUVEC cells.
Collapse
Affiliation(s)
- Jiale Liu
- College of Food Science and Engineering, National Engineering Laboratory of Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Wentian Song
- College of Food Science and Engineering, National Engineering Laboratory of Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Xue Gao
- College of Food Science and Engineering, National Engineering Laboratory of Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Jiaoyan Sun
- College of Food Science and Engineering, National Engineering Laboratory of Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Chunlei Liu
- College of Food Science and Engineering, National Engineering Laboratory of Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Li Fang
- College of Food Science and Engineering, National Engineering Laboratory of Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Ji Wang
- College of Food Science and Engineering, National Engineering Laboratory of Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Junhua Shi
- College of Food Science and Engineering, National Engineering Laboratory of Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Yue Leng
- College of Food Science and Engineering, National Engineering Laboratory of Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Xiaoting Liu
- College of Food Science and Engineering, National Engineering Laboratory of Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun 130118, Jilin, China.
| | - Weihong Min
- College of Food Science and Engineering, National Engineering Laboratory of Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun 130118, Jilin, China; College of Food and Health, Zhejiang A&F University, Hangzhou 311300, China.
| |
Collapse
|
26
|
Al Suleimani YM, Ali BH, Ali H, Manoj P, Almashaiki KS, Abdelrahman AM. The Salutary Effects of Diminazene, Lisinopril or Valsartan on Cisplatin - Induced Acute Kidney Injury in Rats: A Comparative Study. Physiol Res 2024; 73:227-237. [PMID: 38710058 PMCID: PMC11081186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/09/2023] [Indexed: 05/08/2024] Open
Abstract
Nephrotoxicity as a cause of acute kidney injury (AKI) induced by cisplatin (CP), limits its usefulness as an anticancer agent. Diminazene, an angiotensin converting enzyme 2 activator, exhibited renoprotective properties on rat models of kidney diseases. This research aims to investigate the salutary effect of diminazene in comparison with lisinopril or valsartan in CP-induced AKI. The first and second groups of rats received oral vehicle (distilled water) for 9 days, and saline injection or intraperitoneal CP (6 mg/kg) on day 6, respectively. Third, fourth, and fifth groups received intraperitoneal injections of CP on day 6 and diminazene (15 mg/kg/day, orally), lisinopril (10 mg/kg/day, orally), or valsartan (30 mg/kg/day, orally), for 9 days, respectively. 24h after the last day of treatment, blood and kidneys were removed under anesthesia for biochemical and histopathological examination. Urine during the last 24 h before sacrificing the rats was also collected. CP significantly increased plasma urea, creatinine, neutrophil gelatinase-associated lipocalin, calcium, phosphorus, and uric acid. It also increased urinary albumin/creatinine ratio, N-Acetyl-beta-D-Glucosaminidase/creatinine ratio, and reduced creatinine clearance, as well the plasma concentrations of inflammatory cytokines [plasma tumor necrosis factor-alpha, and interleukin-1beta], and significantly reduced antioxidant indices [catalase, glutathione reductase , and superoxide dismutase]. Histopathologically, CP treatment caused necrosis of renal tubules, tubular casts, shrunken glomeruli, and increased renal fibrosis. Diminazine, lisinopril, and valsartan ameliorated CP-induced biochemical and histopathological changes to a similar extent. The salutary effect of the three drugs used is, at least partially, due to their anti-inflammatory and antioxidant effects. Keywords: Cisplatin, Diminazene, ACE2 activator, Lisinopril, Valsartan, Acute kidney injury.
Collapse
Affiliation(s)
- Y M Al Suleimani
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Al Khod, Oman,
| | | | | | | | | | | |
Collapse
|
27
|
Pillai AT, Morya S, Kasankala LM. Emerging Trends in Bioavailability and Pharma-Nutraceutical Potential of Whey Bioactives. J Nutr Metab 2024; 2024:8455666. [PMID: 38633607 PMCID: PMC11023716 DOI: 10.1155/2024/8455666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/18/2023] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
Whey, a component of milk and a useful by-product of the dairy industry's casein and cheese-making, has been used for generations to augment animal feed. It contains a range of proteins, including α-lactalbumin, β-lactoglobulin, bovine serum albumin, heavy and light chain immunoglobulins, lactoferrin, glycomacropeptide, and lactoperoxidase. Whey proteins exhibit great potential as biopolymers for creating bioactive delivery systems owing to their distinct health-enhancing characteristics and the presence of numerous amino acid groups within their structures. Whey has considerable factors such as antitumor, anti-inflammatory, antihypertensive, hypolipidemic, antiviral, and antibacterial properties in addition to chelating. The global market of whey protein stood at USD 5.33 billion in 2021, with a projected compound annual growth rate of 10.48% spanning the interval from 2022 to 2030. The escalating demand for whey protein is intrinsically linked to the amplifying consciousness surrounding healthy lifestyles. Notably, protein supplements are recurrently endorsed by fitness and sports establishments, thereby accentuating the focal point of customers toward whey protein. This review focuses on nutritional composition, whey bioactives, and their bioavailability with potential health benefits.
Collapse
Affiliation(s)
- Adhithyan T. Pillai
- Department of Food Technology and Nutrition, School of Agriculture, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Sonia Morya
- Department of Food Technology and Nutrition, School of Agriculture, Lovely Professional University, Phagwara 144411, Punjab, India
| | | |
Collapse
|
28
|
Weissman D, Dudek J, Sequeira V, Maack C. Fabry Disease: Cardiac Implications and Molecular Mechanisms. Curr Heart Fail Rep 2024; 21:81-100. [PMID: 38289538 PMCID: PMC10923975 DOI: 10.1007/s11897-024-00645-1] [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] [Accepted: 01/05/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE OF REVIEW This review explores the interplay among metabolic dysfunction, oxidative stress, inflammation, and fibrosis in Fabry disease, focusing on their potential implications for cardiac involvement. We aim to discuss the biochemical processes that operate in parallel to sphingolipid accumulation and contribute to disease pathogenesis, emphasizing the importance of a comprehensive understanding of these processes. RECENT FINDINGS Beyond sphingolipid accumulation, emerging studies have revealed that mitochondrial dysfunction, oxidative stress, and chronic inflammation could be significant contributors to Fabry disease and cardiac involvement. These factors promote cardiac remodeling and fibrosis and may predispose Fabry patients to conduction disturbances, ventricular arrhythmias, and heart failure. While current treatments, such as enzyme replacement therapy and pharmacological chaperones, address disease progression and symptoms, their effectiveness is limited. Our review uncovers the potential relationships among metabolic disturbances, oxidative stress, inflammation, and fibrosis in Fabry disease-related cardiac complications. Current findings suggest that beyond sphingolipid accumulation, other mechanisms may significantly contribute to disease pathogenesis. This prompts the exploration of innovative therapeutic strategies and underscores the importance of a holistic approach to understanding and managing Fabry disease.
Collapse
Affiliation(s)
- David Weissman
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Jan Dudek
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Vasco Sequeira
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany.
| |
Collapse
|
29
|
Picod A, Placier S, Genest M, Callebert J, Julian N, Zalc M, Assad N, Nordin H, Santos K, Gaudry S, Chatziantoniou C, Mebazaa A, Azibani F. Circulating Dipeptidyl Peptidase 3 Modulates Systemic and Renal Hemodynamics Through Cleavage of Angiotensin Peptides. Hypertension 2024; 81:927-935. [PMID: 38334001 PMCID: PMC10956665 DOI: 10.1161/hypertensionaha.123.21913] [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: 08/14/2023] [Accepted: 01/25/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND High circulating DPP3 (dipeptidyl peptidase 3) has been associated with poor prognosis in critically ill patients with circulatory failure. In such situation, DPP3 could play a pathological role, putatively via an excessive angiotensin peptides cleavage. Our objective was to investigate the hemodynamics changes induced by DPP3 in mice and the relation between the observed effects and renin-angiotensin system modulation. METHODS Ten-week-old male C57Bl/6J mice were subjected to intravenous injection of purified human DPP3 or an anti-DPP3 antibody (procizumab). Invasive blood pressure and renal blood flow were monitored throughout the experiments. Circulating angiotensin peptides and catecholamines were measured and receptor blocking experiment performed to investigate the underlying mechanisms. RESULTS DPP3 administration significantly increased renal blood flow, while blood pressure was minimally affected. Conversely, procizumab led to significantly decreased renal blood flow. Angiotensin peptides measurement and an AT1R (angiotensin II receptor type 1) blockade experiment using valsartan demonstrated that the renovascular effect induced by DPP3 is due to reduced AT1R activation via decreased concentrations of circulating angiotensin II, III, and IV. Measurements of circulating catecholamines and an adrenergic receptor blockade by labetalol demonstrated a concomitant catecholamines release that explains blood pressure maintenance upon DPP3 administration. CONCLUSIONS High circulating DPP3 increases renal blood flow due to reduced AT1R activation via decreased concentrations of circulating angiotensin peptides while blood pressure is maintained by concomitant endogenous catecholamines release.
Collapse
Affiliation(s)
- Adrien Picod
- Institut National de la Santé et de la Recherche Médicale UMR-S 942 MASCOT – Paris – Cité University, Paris, France (A.P., M.G., J.C., N.J., M.Z., N.A., H.N., A.M., F.A.)
| | - Sandrine Placier
- INSERM UMR-S 1155 CORAKID – Paris – Sorbonne University, France (S.P., S.G., C.C.)
| | - Magali Genest
- Institut National de la Santé et de la Recherche Médicale UMR-S 942 MASCOT – Paris – Cité University, Paris, France (A.P., M.G., J.C., N.J., M.Z., N.A., H.N., A.M., F.A.)
| | - Jacques Callebert
- Institut National de la Santé et de la Recherche Médicale UMR-S 942 MASCOT – Paris – Cité University, Paris, France (A.P., M.G., J.C., N.J., M.Z., N.A., H.N., A.M., F.A.)
- Department of Biochemistry and Molecular Biology, Lariboisière – Saint Louis Hospitals (J.C.), APHP, Paris, France
| | - Nathan Julian
- Institut National de la Santé et de la Recherche Médicale UMR-S 942 MASCOT – Paris – Cité University, Paris, France (A.P., M.G., J.C., N.J., M.Z., N.A., H.N., A.M., F.A.)
- Department of Anesthesiology and Intensive Care, Lariboisière – Saint Louis Hospitals (N.J., A.M.), APHP, Paris, France
| | - Maxime Zalc
- Institut National de la Santé et de la Recherche Médicale UMR-S 942 MASCOT – Paris – Cité University, Paris, France (A.P., M.G., J.C., N.J., M.Z., N.A., H.N., A.M., F.A.)
- Department of Anesthesiology and Intensive Care, Mondor Hospital (M.Z.), APHP, Paris, France
- Paris Est – Créteil University, France (M.Z.)
| | - Noma Assad
- Institut National de la Santé et de la Recherche Médicale UMR-S 942 MASCOT – Paris – Cité University, Paris, France (A.P., M.G., J.C., N.J., M.Z., N.A., H.N., A.M., F.A.)
| | - Hugo Nordin
- Institut National de la Santé et de la Recherche Médicale UMR-S 942 MASCOT – Paris – Cité University, Paris, France (A.P., M.G., J.C., N.J., M.Z., N.A., H.N., A.M., F.A.)
| | - Karine Santos
- 4TEEN4 Pharmaceuticals Gmbh, Hennigsdorf, Germany (K.S.)
| | - Stéphane Gaudry
- INSERM UMR-S 1155 CORAKID – Paris – Sorbonne University, France (S.P., S.G., C.C.)
- Sorbonne – Paris Nord University, France (S.G.)
- Medical and Surgical Intensive Care Unit, Avicenne Hospital, APHP, Bobigny, France (S.G.)
| | | | - Alexandre Mebazaa
- Institut National de la Santé et de la Recherche Médicale UMR-S 942 MASCOT – Paris – Cité University, Paris, France (A.P., M.G., J.C., N.J., M.Z., N.A., H.N., A.M., F.A.)
- Department of Anesthesiology and Intensive Care, Lariboisière – Saint Louis Hospitals (N.J., A.M.), APHP, Paris, France
| | - Feriel Azibani
- Institut National de la Santé et de la Recherche Médicale UMR-S 942 MASCOT – Paris – Cité University, Paris, France (A.P., M.G., J.C., N.J., M.Z., N.A., H.N., A.M., F.A.)
| |
Collapse
|
30
|
Busse LW, Schaich CL, Chappell MC, McCurdy MT, Staples EM, Ten Lohuis CC, Hinson JS, Sevransky JE, Rothman RE, Wright DW, Martin GS, Khanna AK. Association of Active Renin Content With Mortality in Critically Ill Patients: A Post hoc Analysis of the Vitamin C, Thiamine, and Steroids in Sepsis (VICTAS) Trial. Crit Care Med 2024; 52:441-451. [PMID: 37947484 PMCID: PMC10876175 DOI: 10.1097/ccm.0000000000006095] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
OBJECTIVE Sepsis is a leading cause of mortality. Predicting outcomes is challenging and few biomarkers perform well. Defects in the renin-angiotensin system (RAS) can predict clinical outcomes in sepsis and may outperform traditional biomarkers. We postulated that RAS dysfunction (elevated active renin, angiotensin 1-7 [Ang-(1-7)], and angiotensin-converting enzyme 2 (ACE2) activity with depressed Ang-II and ACE activity) would be associated with mortality in a cohort of septic patients. DESIGN Post hoc analysis of patients enrolled in the Vitamin C, Thiamine, and Steroids in Sepsis (VICTAS) randomized controlled trial. SETTING Forty-three hospitals across the United States. PATIENTS Biorepository samples of 103 patients. INTERVENTIONS We analyzed day 0 (within 24 hr of respiratory failure, septic shock, or both) and day 3 samples ( n = 103 and 95, respectively) for assessment of the RAS. The association of RAS values with 30-day mortality was determined using Cox proportional hazards regression with multivariable adjustments for age, sex, VICTAS treatment arm, systolic blood pressure, Sequential Organ Failure Assessment Score, and vasopressor use. MEASUREMENTS AND MAIN RESULTS High baseline active renin values were associated with higher 30-day mortality when dichotomized to the median of 188.7 pg/mL (hazard ratio [HR] = 2.84 [95% CI, 1.10-7.33], p = 0.031) or stratified into quartiles (Q1 = ref, HR Q2 = 2.01 [0.37-11.04], HR Q3 = 3.22 [0.64-16.28], HR Q4 = 5.58 [1.18-26.32], p for linear trend = 0.023). A 1- sd (593.6 pg/mL) increase in renin from day 0 to day 3 was associated with increased mortality (HR = 3.75 [95% CI, 1.94-7.22], p < 0.001), and patients whose renin decreased had improved survival compared with those whose renin increased (HR 0.22 [95% CI, 0.08-0.60], p = 0.003). Ang-(1-7), ACE2 activity, Ang-II and ACE activity did not show this association. Mortality was attenuated in patients with renin over the median on day 0 who received the VICTAS intervention, but not on day 3 ( p interaction 0.020 and 0.137, respectively). There were no additional consistent patterns of mortality on the RAS from the VICTAS intervention. CONCLUSIONS Baseline serum active renin levels were strongly associated with mortality in critically ill patients with sepsis. Furthermore, a greater relative activation in circulating renin from day 0 to day 3 was associated with a higher risk of death.
Collapse
Affiliation(s)
- Laurence W Busse
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA
- Emory Critical Care Center, Emory Healthcare, Atlanta, GA
| | - Christopher L Schaich
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Mark C Chappell
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Michael T McCurdy
- Division of Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Erin M Staples
- Department of Anesthesiology, Section on Critical Care Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | | | - Jeremiah S Hinson
- Department of Emergency Medicine, Johns Hopkins School of Medicine, Baltimore, MD
| | - Jonathan E Sevransky
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA
- Emory Critical Care Center, Emory Healthcare, Atlanta, GA
| | - Richard E Rothman
- Department of Emergency Medicine, Johns Hopkins University, The Johns Hopkins Hospital, Baltimore, MD
| | - David W Wright
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA
- Grady Marcus Trauma and Emergency Care Center, Atlanta, GA
| | - Greg S Martin
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA
- Emory Critical Care Center, Emory Healthcare, Atlanta, GA
| | - Ashish K Khanna
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA
- Emory Critical Care Center, Emory Healthcare, Atlanta, GA
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC
- Division of Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore, MD
- Department of Anesthesiology, Section on Critical Care Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
- Department of Emergency Medicine, Johns Hopkins School of Medicine, Baltimore, MD
- Department of Emergency Medicine, Johns Hopkins University, The Johns Hopkins Hospital, Baltimore, MD
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA
- Grady Marcus Trauma and Emergency Care Center, Atlanta, GA
- Department of Anesthesiology, Section of Critical Care Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
- Outcomes Research Consortium, Cleveland, OH
- Perioperative Outcomes and Informatics Collaborative, Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC
| |
Collapse
|
31
|
Lane A, Jiles M, Ramey K, McLean M, Whitney J, Brunson A, Cardaci T, Liu J, Wilcox S, Catov J, Fernhall B. Adverse pregnancy outcomes and renal-vascular function in the early years after delivery. Am J Physiol Heart Circ Physiol 2024; 326:H82-H88. [PMID: 37921666 PMCID: PMC11555665 DOI: 10.1152/ajpheart.00641.2023] [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: 10/06/2023] [Accepted: 10/31/2023] [Indexed: 11/04/2023]
Abstract
Adverse pregnancy outcomes (APOs: hypertensive disorders, gestational diabetes, preterm birth, and placental disorders) are associated with cardiovascular disease risk or blood volume abnormalities. Traditional risk factors might not identify highest risk people in the early years after APO deliveries. Test the hypothesis that vascular function is worse, and plasma volume-regulating renal hormones are lower after delivery in people who did versus did not have an APO. Adult participants 6 mo-3 years postdelivery of a singleton infant participated in this cross-sectional study. Exclusion criteria included current smoking, current use of certain medications, and diabetes outside of pregnancy. Differences in measurements between participants with versus without APOs were determined with t tests or Wilcoxon tests. Associations of renal hormones with APO history were assessed with linear regression, adjusted for age, race, body mass index (BMI), and sodium consumption. Of 86 participants, 38 (44%) had an APO history. Those with APOs were more likely to identify as Black and had a higher BMI, 34.0 kg/m2 [interquartile range (IQR), 24.6, 39.3] versus 24.2 kg/m2 [IQR, 21.2, 31.3], P < 0.05. Most brachial and all aortic blood pressures were higher in those with APOs: median aortic blood pressure was 102/74 versus 96/68 mmHg, P ≤ 0.05. There were no differences in arterial stiffness or endothelial function between groups. Aldosterone was lower (54 [IQR, 28-84] vs. 80 [IQR, 39-150] pmol/L) in participants with past APOs. Blood pressures were higher, and aldosterone was lower in participants with past APOs. Associations of aldosterone with APO history persisted after adjustment. Neither renin nor aldosterone were related to vascular function.NEW & NOTEWORTHY Adverse pregnancy outcomes (APOs) are associated with cardiovascular disease (CVD) risk. Traditional CVD risk factors may not fully capture excess CVD risk soon after APOs. Vascular dysfunction and plasma volume irregularities may be detectable. We found people with APOs had worse blood pressures, higher BMI, and lower aldosterone levels versus those without APOs in the early years after delivery. Vascular function was similar between groups. Future research should assess vascular function and renal hormones at multiple timepoints during the perinatal period.
Collapse
Affiliation(s)
- Abbi Lane
- Department of Applied Exercise Science, School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Marcey Jiles
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Kaitlyn Ramey
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Marnie McLean
- Department of Applied Exercise Science, School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Jaime Whitney
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Abigail Brunson
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Thomas Cardaci
- Department of Immunology, School of Medicine, University of South Carolina, Columbia, SC
| | - Jihong Liu
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Sara Wilcox
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Janet Catov
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA
- Department of Obstetrics and Gynecology, Magee-Women’s Research Institute, Pittsburgh, PA
| | - Bo Fernhall
- Manning College of Nursing and Health Sciences, University of Massachusetts Boston, Boston, MA
| |
Collapse
|
32
|
Demeuse J, Huyghebaert L, Determe W, Schoumacher M, Grifnée E, Massonnet P, Dubrowski T, Rechchad M, Segura JF, Peeters S, Cavalier E, Le Goff C. Development and validation of an LC-MS/MS method for the simultaneous quantitation of angiotensin (1-7), (1-8), (1-9) and (1-10) in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1232:123943. [PMID: 38039597 DOI: 10.1016/j.jchromb.2023.123943] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/03/2023]
Abstract
Cardiovascular diseases have cast a significant negative impact on the lives of millions worldwide. Over the years, extensive efforts have been dedicated to enhancing diagnostic and prognostic tools for these diseases. A growing body of evidence indicates that the angiotensin convertase enzyme (ACE) and the angiotensin convertase enzyme 2 (ACE2), and angiotensin peptide levels could hold a pivotal role in assisting clinicians with the management of cardiovascular conditions, notably hypertension and heart failure. However, despite the considerable body of knowledge in this domain, a void remains in the field of analytical methodologies for these molecules. In this study, we present a fully validated LC-MS/MS method for the precise quantitation of plasma angiotensin (1-7), (1-8), (1-9), and (1-10), following the guidelines set by the Clinical and Laboratory Standards Institute (CLSI). Our method not only enables the accurate quantification of angiotensin peptides but also provides a means to assess ACE and ACE2 activity. Remarkably, our method achieved a Lower Limit of Measurement Interval (LLMI) as low as 5 pg/mL. This has enabled the detection of angiotensin (1-7), (1-8), (1-9) and (1-10) and the accurate quantitation of angiotensin (1-7), (1-8) and (1-10) in all analyzed groups, including healthy controls, patients with high blood pressure, and patients with chronic kidney disease. To our knowledge, our method represents the most sensitive approach allowing for simultaneous quantitation of these four angiotensin peptides. A distinct advantage of our method, when compared to immunoassays, is its high sensitivity combined with comprehensive chromatographic separation of all currently known angiotensin peptides. This combination translates to an exceptional level of selectivity, underscoring the value and potential of our methodology in advancing cardiovascular disease research.
Collapse
Affiliation(s)
- Justine Demeuse
- Department of Clinical Chemistry, CIRM, University of Liège, Belgium
| | - Loreen Huyghebaert
- Department of Clinical Chemistry, University Hospital of Liège, Belgium.
| | - William Determe
- Department of Clinical Chemistry, CIRM, University of Liège, Belgium
| | | | - Elodie Grifnée
- Department of Clinical Chemistry, University Hospital of Liège, Belgium
| | | | - Thomas Dubrowski
- Department of Clinical Chemistry, University Hospital of Liège, Belgium
| | - Marwa Rechchad
- Department of Clinical Chemistry, University Hospital of Liège, Belgium
| | | | - Stéphanie Peeters
- Department of Clinical Chemistry, University Hospital of Liège, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, CIRM, University of Liège, Belgium; Department of Clinical Chemistry, University Hospital of Liège, Belgium
| | - Caroline Le Goff
- Department of Clinical Chemistry, CIRM, University of Liège, Belgium; Department of Clinical Chemistry, University Hospital of Liège, Belgium
| |
Collapse
|
33
|
Garcia B, Zarbock A, Bellomo R, Legrand M. The alternative renin-angiotensin system in critically ill patients: pathophysiology and therapeutic implications. Crit Care 2023; 27:453. [PMID: 37986086 PMCID: PMC10662652 DOI: 10.1186/s13054-023-04739-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023] Open
Abstract
The renin-angiotensin system (RAS) plays a crucial role in regulating blood pressure and the cardio-renal system. The classical RAS, mainly mediated by angiotensin I, angiotensin-converting enzyme, and angiotensin II, has been reported to be altered in critically ill patients, such as those in vasodilatory shock. However, recent research has highlighted the role of some components of the counterregulatory axis of the classical RAS, termed the alternative RAS, such as angiotensin-converting Enzyme 2 (ACE2) and angiotensin-(1-7), or peptidases which can modulate the RAS like dipeptidyl-peptidase 3, in many critical situations. In cases of shock, dipeptidyl-peptidase 3, an enzyme involved in the degradation of angiotensin and opioid peptides, has been associated with acute kidney injury and mortality and preclinical studies have tested its neutralization. Angiotensin-(1-7) has been shown to prevent septic shock development and improve outcomes in experimental models of sepsis. In the context of experimental acute lung injury, ACE2 activity has demonstrated a protective role, and its inactivation has been associated with worsened lung function, leading to the use of active recombinant human ACE2, in preclinical and human studies. Angiotensin-(1-7) has been tested in experimental models of acute lung injury and in a recent randomized controlled trial for patients with COVID-19 related hypoxemia. Overall, the alternative RAS appears to have a role in the pathogenesis of disease in critically ill patients, and modulation of the alternative RAS may improve outcomes. Here, we review the available evidence regarding the methods of analysis of the RAS, pathophysiological disturbances of this system, and discuss how therapeutic manipulation may improve outcomes in the critically ill.
Collapse
Affiliation(s)
- Bruno Garcia
- Department of Anesthesia and Peri-Operative Care, Division of Critical Care Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Department of Intensive Care, Centre Hospitalier Universitaire de Lille, Lille, France
- Experimental Laboratory of the Department of Intensive Care, Université Libre de Bruxelles, Brussels, Belgium
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, Münster, Germany
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Hospital, Melbourne, VIC, 3084, Australia
- Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, Australia
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, VIC, Australia
| | - Matthieu Legrand
- Department of Anesthesia and Peri-Operative Care, Division of Critical Care Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA.
| |
Collapse
|
34
|
Hung WT, Sutopo CCY, Wu ML, Hsu JL. Discovery and Characterization of a Dual-Function Peptide Derived from Bitter Gourd Seed Protein Using Two Orthogonal Bioassay-Guided Fractionations Coupled with In Silico Analysis. Pharmaceuticals (Basel) 2023; 16:1629. [PMID: 38004494 PMCID: PMC10674851 DOI: 10.3390/ph16111629] [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: 10/07/2023] [Revised: 11/04/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
The hydrolysate of bitter gourd seed protein, digested by the combined gastrointestinal proteases (BGSP-GPs), exhibited the most potent inhibition on angiotensin-I-converting enzyme (ACE) with an IC50 value of 48.1 ± 2.0 µg/mL. Using two independent bioassay-guided fractionations, fraction F5 from reversed-phase chromatography and fraction S1 from strong cation exchange chromatography exhibited the highest ACE inhibitory (ACEI) activity. Three identical peptides were simultaneously detected from both fractions and, based on the in silico appraisal, APLVSW (AW6) was predicted as a promising ACEI peptide. Their dipeptidyl peptidase-IV (DPP4) inhibitory (DPP4I) activity was also explored. The IC50 values of AW6 against ACE and DPP4 were calculated to be 9.6 ± 0.3 and 145.4 ± 4.4 µM, respectively. The inhibitory kinetics and intermolecular interaction studies suggested that AW6 is an ACE competitive inhibitor and a DPP4 non-competitive inhibitor. The quantities of AW6 in BGSP-GP hydrolysate, fractions F5 and S1, were also analyzed using liquid chromatography-tandem mass spectrometry. Notably, AW6 could resist hydrolysis in the human gastrointestinal tract according to the result of the simulated gastrointestinal digestion. To the best of our knowledge, this is the first discovery and characterization of a dual-function (ACEI and DPP4I activities) peptide derived from bitter gourd seed protein.
Collapse
Affiliation(s)
- Wei-Ting Hung
- Department of Food Science, National Pingtung University of Science and Technology, Pingtung 912, Taiwan; (W.-T.H.); (M.-L.W.)
| | - Christoper Caesar Yudho Sutopo
- Department of Tropical Agriculture and International Cooperation, National Pingtung University of Science and Technology, Pingtung 912, Taiwan;
| | - Mei-Li Wu
- Department of Food Science, National Pingtung University of Science and Technology, Pingtung 912, Taiwan; (W.-T.H.); (M.-L.W.)
| | - Jue-Liang Hsu
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| |
Collapse
|
35
|
Ames MK, Adin DB, Wood J. Beyond Angiotensin-Converting Enzyme Inhibitors: Modulation of the Renin-Angiotensin-Aldosterone System to Delay or Manage Congestive Heart Failure. Vet Clin North Am Small Anim Pract 2023; 53:1353-1366. [PMID: 37423846 DOI: 10.1016/j.cvsm.2023.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
The renin-angiotensin-aldosterone system (RAAS) consists of bioactive angiotensin peptides, enzymatic pathways, receptors, and the steroid hormone aldosterone. The RAAS regulates blood pressure, sodium, and electrolyte homeostasis and mediates pathologic disease processes. Within this system is an alternative arm that counterbalances the vasoconstrictive, sodium and water retentive, and pro-fibrotic and inflammatory effects of the classical arm. Improved biochemical methodologies in RAAS quantification are elucidating how this complex system changes in health and disease. Future treatments for cardiovascular and kidney disease will likely involve a more nuanced manipulation of this system rather than simple blockade.
Collapse
Affiliation(s)
- Marisa K Ames
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California - Davis, 1 Shields Avenue, Davis, CA 95616, USA.
| | - Darcy B Adin
- University of Florida, College of Veterinary Medicine, 2015 Southwest 16th Avenue, Gainesville, FL 32608, USA
| | - James Wood
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California - Davis, 1 Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
36
|
Villapol S, Janatpour ZC, Affram KO, Symes AJ. The Renin Angiotensin System as a Therapeutic Target in Traumatic Brain Injury. Neurotherapeutics 2023; 20:1565-1591. [PMID: 37759139 PMCID: PMC10684482 DOI: 10.1007/s13311-023-01435-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health problem, with limited pharmacological options available beyond symptomatic relief. The renin angiotensin system (RAS) is primarily known as a systemic endocrine regulatory system, with major roles controlling blood pressure and fluid homeostasis. Drugs that target the RAS are used to treat hypertension, heart failure and kidney disorders. They have now been used chronically by millions of people and have a favorable safety profile. In addition to the systemic RAS, it is now appreciated that many different organ systems, including the brain, have their own local RAS. The major ligand of the classic RAS, Angiotensin II (Ang II) acts predominantly through the Ang II Type 1 receptor (AT1R), leading to vasoconstriction, inflammation, and heightened oxidative stress. These processes can exacerbate brain injuries. Ang II receptor blockers (ARBs) are AT1R antagonists. They have been shown in several preclinical studies to enhance recovery from TBI in rodents through improvements in molecular, cellular and behavioral correlates of injury. ARBs are now under consideration for clinical trials in TBI. Several different RAS peptides that signal through receptors distinct from the AT1R, are also potential therapeutic targets for TBI. The counter regulatory RAS pathway has actions that oppose those stimulated by AT1R signaling. This alternative pathway has many beneficial effects on cells in the central nervous system, bringing about vasodilation, and having anti-inflammatory and anti-oxidative stress actions. Stimulation of this pathway also has potential therapeutic value for the treatment of TBI. This comprehensive review will provide an overview of the various components of the RAS, with a focus on their direct relevance to TBI pathology. It will explore different therapeutic agents that modulate this system and assess their potential efficacy in treating TBI patients.
Collapse
Affiliation(s)
- Sonia Villapol
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
| | - Zachary C Janatpour
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kwame O Affram
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Aviva J Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
37
|
Stanski NL, Pode Shakked N, Zhang B, Cvijanovich NZ, Fitzgerald JC, Jain PN, Schwarz AJ, Nowak J, Weiss SL, Allen GL, Thomas NJ, Haileselassie B, Goldstein SL. Serum renin and prorenin concentrations predict severe persistent acute kidney injury and mortality in pediatric septic shock. Pediatr Nephrol 2023; 38:3099-3108. [PMID: 36939916 PMCID: PMC10588759 DOI: 10.1007/s00467-023-05930-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 03/21/2023]
Abstract
BACKGROUND Studies in critically ill adults demonstrate associations between serum renin concentrations (a proposed surrogate for renin-angiotensin-aldosterone system dysregulation) and poor outcomes, but data in critically ill children are lacking. We assessed serum renin + prorenin concentrations in children with septic shock to determine their predictive ability for acute kidney injury (AKI) and mortality. METHODS We conducted a secondary analysis of a multicenter observational study of children aged 1 week to 18 years admitted to 14 pediatric intensive care units (PICUs) with septic shock and residual serum available for renin + prorenin measurement. Primary outcomes were development of severe persistent AKI (≥ KDIGO stage 2 for ≥ 48 h) in the first week and 28-day mortality. RESULTS Among 233 patients, day 1 median renin + prorenin concentration was 3436 pg/ml (IQR 1452-6567). Forty-two (18%) developed severe persistent AKI and 32 (14%) died. Day 1 serum renin + prorenin predicted severe persistent AKI with an AUROC of 0.75 (95% CI 0.66-0.84, p < 0.0001; optimal cutoff 6769 pg/ml) and mortality with an AUROC of 0.79 (95% CI 0.69-0.89, p < 0.0001; optimal cutoff 6521 pg/ml). Day 3/day 1 (D3:D1) renin + prorenin ratio had an AUROC of 0.73 (95% CI 0.63-0.84, p < 0.001) for mortality. On multivariable regression, day 1 renin + prorenin > optimal cutoff retained associations with severe persistent AKI (aOR 6.8, 95% CI 3.0-15.8, p < 0.001) and mortality (aOR 6.9, 95% CI 2.2-20.9, p < 0.001). Similarly, D3:D1 renin + prorenin > optimal cutoff was associated with mortality (aOR 7.6, 95% CI 2.5-23.4, p < 0.001). CONCLUSIONS Children with septic shock have very elevated serum renin + prorenin concentrations on PICU admission, and these concentrations, as well as their trend over the first 72 h, predict severe persistent AKI and mortality. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Natalja L Stanski
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45267, USA.
| | - Naomi Pode Shakked
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Bin Zhang
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | | | - Julie C Fitzgerald
- The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Parag N Jain
- Texas Children's Hospital and Baylor College of Medicine, 6621 Fannin Street, Houston, TX, 77030, USA
| | - Adam J Schwarz
- Children's Hospital of Orange County, 1201 W La Veta Ave, Orange, CA, 92868, USA
| | - Jeffrey Nowak
- Children's Minnesota, 2525 Chicago Ave, Minneapolis, MN, 55404, USA
| | - Scott L Weiss
- The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Geoffrey L Allen
- Children's Mercy Hospital, 2401 Gillham Road, Kansas City, MO, 64108, USA
| | - Neal J Thomas
- Penn State Health Children's Hospital, 600 University Dr, Hershey, PA, 17033, USA
| | | | - Stuart L Goldstein
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45267, USA
| |
Collapse
|
38
|
Shoemaker R, Poglitsch M, Huang H, Vignes K, Srinivasan A, Cockerham C, Schadler A, Bauer JA, O’Brien JM. Activation of the Renin-Angiotensin-Aldosterone System Is Attenuated in Hypertensive Compared with Normotensive Pregnancy. Int J Mol Sci 2023; 24:12728. [PMID: 37628909 PMCID: PMC10454898 DOI: 10.3390/ijms241612728] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Hypertension during pregnancy increases the risk of adverse maternal and fetal outcomes, but the mechanisms of pregnancy hypertension are not precisely understood. Elevated plasma renin activity and aldosterone concentrations play an important role in the normal physiologic adaptation to pregnancy. These effectors are reduced in patients with pregnancy hypertension, creating an opportunity to define the features of the renin-angiotensin-aldosterone system (RAAS) that are characteristic of this disorder. In the current study, we used a novel LC-MS/MS-based methodology to develop comprehensive profiles of RAAS peptides and effectors over gestation in a cohort of 74 pregnant women followed prospectively for the development of gestational hypertension and pre-eclampsia (HYP, 27 patients) versus those remaining normotensive (NT, 47 patients). In NT pregnancy, the plasma renin activity surrogate, (PRA-S, calculated from the sum of Angiotensin I + Angiotensin II) and aldosterone concentrations significantly increased from the first to the third trimester, accompanied by a modest increase in the concentrations of angiotensin peptide metabolites. In contrast, in HYP pregnancies, PRA-S and angiotensin peptides were largely unchanged over gestation, and third-trimester aldosterone concentrations were significantly lower compared with those in NT pregnancies. The results indicated that the predominant features of pregnancies that develop HYP are stalled or waning activation of the RAAS in the second half of pregnancy (accompanied by unchanging levels of angiotensin peptides) and the attenuated secretion of aldosterone.
Collapse
Affiliation(s)
- Robin Shoemaker
- Department of Dietetics and Human Nutrition, University of Kentucky, Lexington, KY 40506, USA
| | | | - Hong Huang
- Department of Pediatrics, University of Kentucky, Lexington, KY 40536, USA
| | - Katherine Vignes
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY 40506, USA
| | - Aarthi Srinivasan
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY 40506, USA
| | - Cynthia Cockerham
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY 40506, USA
| | - Aric Schadler
- Department of Pediatrics, University of Kentucky, Lexington, KY 40536, USA
| | - John A. Bauer
- Department of Pediatrics, University of Kentucky, Lexington, KY 40536, USA
| | - John M. O’Brien
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
39
|
Visniauskas B, Kilanowski-Doroh I, Ogola BO, Mcnally AB, Horton AC, Imulinde Sugi A, Lindsey SH. Estrogen-mediated mechanisms in hypertension and other cardiovascular diseases. J Hum Hypertens 2023; 37:609-618. [PMID: 36319856 PMCID: PMC10919324 DOI: 10.1038/s41371-022-00771-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 06/08/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally for men and women. Premenopausal women have a lower incidence of hypertension and other cardiovascular events than men of the same age, but diminished sex differences after menopause implicates 17-beta-estradiol (E2) as a protective agent. The cardioprotective effects of E2 are mediated by nuclear estrogen receptors (ERα and ERβ) and a G protein-coupled estrogen receptor (GPER). This review summarizes both established as well as emerging estrogen-mediated mechanisms that underlie sex differences in the vasculature during hypertension and CVD. In addition, remaining knowledge gaps inherent in the association of sex differences and E2 are identified, which may guide future clinical trials and experimental studies in this field.
Collapse
Affiliation(s)
- Bruna Visniauskas
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Benard O Ogola
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Alexandra B Mcnally
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Alec C Horton
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ariane Imulinde Sugi
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Center of Excellence in Sex-Based Biology and Medicine, New Orleans, LA, USA.
- Tulane Brain Institute, New Orleans, LA, USA.
| |
Collapse
|
40
|
Tibi S, Zeynalvand G, Mohsin H. Role of the Renin Angiotensin Aldosterone System in the Pathogenesis of Sepsis-Induced Acute Kidney Injury: A Systematic Review. J Clin Med 2023; 12:4566. [PMID: 37510681 PMCID: PMC10380384 DOI: 10.3390/jcm12144566] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/06/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Sepsis is a life-threatening condition responsible for up to 20% of all global deaths. Kidneys are among the most common organs implicated, yet the pathogenesis of sepsis-induced acute kidney injury (S-AKI) is not completely understood, resulting in the treatment being nonspecific and responsive. In situations of stress, the renin angiotensin aldosterone system (RAAS) may play a role. This systematic review focuses on analyzing the impact of the RAAS on the development of S-AKI and discussing the use of RAAS antagonists as an emerging therapeutic option to minimize complications of sepsis. METHODS Studies were identified using electronic databases (Medline via PubMed, Google Scholar) published within the past decade, comprised from 2014 to 2023. The search strategy was conducted using the following keywords: sepsis, S-AKI, RAAS, Angiotensin II, and RAAS inhibitors. Studies on human and animal subjects were included if relevant to the keywords. RESULTS Our search identified 22 eligible references pertaining to the inclusion criteria. Treatment of sepsis with RAAS inhibitor medications is observed to decrease rates of S-AKI, reduce the severity of S-AKI, and offer an improved prognosis for septic patients. CONCLUSION The use of RAAS antagonists as a treatment after the onset of sepsis has promising findings, with evidence of decreased renal tissue damage and rates of S-AKI and improved survival outcomes. REGISTRATION INPLASY202360098.
Collapse
Affiliation(s)
- Sedra Tibi
- School of Medicine, California University of Science and Medicine, Colton, CA 92324, USA
| | - Garbel Zeynalvand
- School of Medicine, California University of Science and Medicine, Colton, CA 92324, USA
| | - Hina Mohsin
- School of Medicine, California University of Science and Medicine, Colton, CA 92324, USA
| |
Collapse
|
41
|
Yudho Sutopo CC, Aznam N, Arianingrum R, Hsu JL. Screening potential hypertensive peptides using two consecutive bioassay-guided SPE fractionations and identification of an ACE inhibitory peptide, DHSTAVW (DW7), derived from pearl garlic protein hydrolysate. Peptides 2023; 167:171046. [PMID: 37330111 DOI: 10.1016/j.peptides.2023.171046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
The pearl garlic (Allium sativum L.) protein (PGP) was digested using pepsin, trypsin, α-chymotrypsin, thermolysin, and simulated gastrointestinal digestion. The α-chymotrypsin hydrolysate showed the highest angiotensin-I-converting enzyme inhibitory (ACEI) activity, with an IC50 value of 190.9 ± 11µg/mL. A reversed-phase C18 solid-phase extraction (RP-SPE) cartridge was used for the first fractionation, and the S4 fraction from RP-SPE showed the most potent ACEI activity (IC50 = 124.1 ± 11 3µg/mL). The S4 fraction was further fractionated using a hydrophilic interaction liquid chromatography SPE (HILIC-SPE). The H4 fraction from HILIC-SPE showed the highest ACEI activity (IC50 = 57.7 ± 3µg/mL). Four ACEI peptides (DHSTAVW, KLAKVF, KLSTAASF, and KETPEAHVF) were identified from the H4 fraction using liquid chromatography-tandem mass spectrometry (LC-MS/MS), and their biological activities were appraised in silico. Among the identified α-chymotryptic peptides, DHSTAVW (DW7), derived from I lectin partial protein, exhibited the most potent ACEI activity (IC50 value of 2.8 ± 0.1µM). DW7 was resistant to simulated gastrointestinal digestion, and it was classified as a prodrug-type inhibitor according to the preincubation experiment. The inhibition kinetics indicated that DW7 was a competitive inhibitor, which was rationalized by the molecular docking simulation. The quantities of DW7 in 1mg of hydrolysate, S4 fraction, and H4 fraction were quantified using LC-MS/MS to give 3.1 ± 0.1, 4.2 ± 0.1, and 13.2 ± 0.1µg, respectively. The amount of DW7 was significantly increased by 4.2-fold compared with the hydrolysate, which suggested that this method is efficient for active peptide screening.
Collapse
Affiliation(s)
- Christoper Caesar Yudho Sutopo
- Department of Tropical Agriculture and International Cooperation, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Nurfina Aznam
- Department of Chemistry Education, Faculty of Mathematics and Natural Sciences, Yogyakarta State University, Sleman 55281, Indonesia
| | - Retno Arianingrum
- Department of Chemistry Education, Faculty of Mathematics and Natural Sciences, Yogyakarta State University, Sleman 55281, Indonesia
| | - Jue-Liang Hsu
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan; Institute of Food Safety Management, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan.
| |
Collapse
|
42
|
Abstract
The current epidemic of corona virus disease (COVID-19) has resulted in an immense health burden that became the third leading cause of death and potentially contributed to a decline in life expectancy in the United States. The severe acute respiratory syndrome-related coronavirus-2 binds to the surface-bound peptidase angiotensin-converting enzyme 2 (ACE2, EC 3.4.17.23) leading to tissue infection and viral replication. ACE2 is an important enzymatic component of the renin-angiotensin system (RAS) expressed in the lung and other organs. The peptidase regulates the levels of the peptide hormones Ang II and Ang-(1-7), which have distinct and opposing actions to one another, as well as other cardiovascular peptides. A potential consequence of severe acute respiratory syndrome-related coronavirus-2 infection is reduced ACE2 activity by internalization of the viral-ACE2 complex and subsequent activation of the RAS (higher ratio of Ang II:Ang-[1-7]) that may exacerbate the acute inflammatory events in COVID-19 patients and possibly contribute to the effects of long COVID-19. Moreover, COVID-19 patients present with an array of autoantibodies to various components of the RAS including the peptide Ang II, the enzyme ACE2, and the AT1 AT2 and Mas receptors. Greater disease severity is also evident in male COVID-19 patients, which may reflect underlying sex differences in the regulation of the 2 distinct functional arms of the RAS. The current review provides a critical evaluation of the evidence for an activated RAS in COVID-19 subjects and whether this system contributes to the greater severity of severe acute respiratory syndrome-related coronavirus-2 infection in males as compared with females.
Collapse
Affiliation(s)
- Mark C. Chappell
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
43
|
Alexander BT, South AM, August P, Bertagnolli M, Ferranti EP, Grobe JL, Jones EJ, Loria AS, Safdar B, Sequeira-Lopez MLS. Appraising the Preclinical Evidence of the Role of the Renin-Angiotensin-Aldosterone System in Antenatal Programming of Maternal and Offspring Cardiovascular Health Across the Life Course: Moving the Field Forward: A Scientific Statement From the American Heart Association. Hypertension 2023; 80:e75-e89. [PMID: 36951054 PMCID: PMC10242542 DOI: 10.1161/hyp.0000000000000227] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
There is increasing interest in the long-term cardiovascular health of women with complicated pregnancies and their affected offspring. Emerging antenatal risk factors such as preeclampsia appear to increase the risk of hypertension and cardiovascular disease across the life course in both the offspring and women after pregnancy. However, the antenatal programming mechanisms responsible are complex and incompletely understood, with roots in alterations in the development, structure, and function of the kidney, heart, vasculature, and brain. The renin-angiotensin-aldosterone system is a major regulator of maternal-fetal health through the placental interface, as well as kidney and cardiovascular tissue development and function. Renin-angiotensin-aldosterone system dysregulation plays a critical role in the development of pregnancy complications such as preeclampsia and programming of long-term adverse cardiovascular health in both the mother and the offspring. An improved understanding of antenatal renin-angiotensin-aldosterone system programming is crucial to identify at-risk individuals and to facilitate development of novel therapies to prevent and treat disease across the life course. Given the inherent complexities of the renin-angiotensin-aldosterone system, it is imperative that preclinical and translational research studies adhere to best practices to accurately and rigorously measure components of the renin-angiotensin-aldosterone system. This comprehensive synthesis of preclinical and translational scientific evidence of the mechanistic role of the renin-angiotensin-aldosterone system in antenatal programming of hypertension and cardiovascular disease will help (1) to ensure that future research uses best research practices, (2) to identify pressing needs, and (3) to guide future investigations to maximize potential outcomes. This will facilitate more rapid and efficient translation to clinical care and improve health outcomes.
Collapse
|
44
|
Butts B, Goeddel LA, Zheng J, Pat B, Powell P, Mobley J, Ahmad S, Steele C, McGiffin D, Davies JE, George JF, Melby SJ, Ferrario CM, Dell’Italia LJ. Impact of early pericardial fluid chymase activation after cardiac surgery. Front Cardiovasc Med 2023; 10:1132786. [PMID: 37265571 PMCID: PMC10230304 DOI: 10.3389/fcvm.2023.1132786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/20/2023] [Indexed: 06/03/2023] Open
Abstract
Introduction Chymase is a highly destructive serine protease rapidly neutralized in the circulation by protease inhibitors. Here we test whether pericardial fluid (PCF) chymase activation and other inflammatory biomarkers determine intensive care unit length of stay, and explore mechanisms of chymase delivery by extracellular vesicles to the heart. Methods PCF was collected from adult patients (17 on-pump; 13 off-pump) 4 h after cardiac surgery. Extracellular vesicles (EVs) containing chymase were injected into Sprague-Dawley rats to test for their ability to deliver chymase to the heart. Results The mean intensive care unit (ICU) stay and mean total length of stay was 2.17 ± 3.8 days and 6.41 ± 1.3 days respectively. Chymase activity and 32 inflammatory markers did not differ in on-pump vs. off-pump cardiac surgery. Society of Thoracic Surgeons Predicted Risk of Morbidity and Mortality Score (STS-PROM), 4-hour post-surgery PCF chymase activity and C-X-C motif chemokine ligand 6 (CXCL6) were all independent predictors of ICU and total hospital length of stay by univariate analysis. Mass spectrometry of baseline PCF shows the presence of serine protease inhibitors that neutralize chymase activity. The compartmentalization of chymase within and on the surface of PCF EVs was visualized by immunogold labeling and transmission electron microscopy. A chymase inhibitor prevented EV chymase activity (0.28 fmol/mg/min vs. 14.14 fmol/mg/min). Intravenous injection of PCF EVs obtained 24 h after surgery into Sprague Dawley rats shows diffuse human chymase uptake in the heart with extensive cardiomyocyte damage 4 h after injection. Discussion Early postoperative PCF chymase activation underscores its potential role in cardiac damage soon after on- or off-pump cardiac surgery. In addition, chymase in extracellular vesicles provides a protected delivery mechanism from neutralization by circulating serine protease inhibitors.
Collapse
Affiliation(s)
- Brittany Butts
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States
| | - Lee A. Goeddel
- Department of Anesthesia and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Jingyi Zheng
- Department of Mathematics and Statistics, College of Science and Mathematics, Auburn University, Auburn, AL, United States
| | - Betty Pat
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham (UAB), Birmingham, AL, United States
- Department of Veterans Affairs, Birmingham Veterans Affairs Health Care System, Birmingham, AL, United States
| | - Pamela Powell
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham (UAB), Birmingham, AL, United States
- Department of Veterans Affairs, Birmingham Veterans Affairs Health Care System, Birmingham, AL, United States
| | - James Mobley
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
| | - Sarfaraz Ahmad
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Chad Steele
- School of Medicine—Microbiology and Immunology, Tulane University, New Orleans, LA, United States
| | - David McGiffin
- Cardiothoracic Surgery and Transplantation, The Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - James E. Davies
- Department of Surgery, Division of Cardiothoracic Surgery, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
| | - James F. George
- Department of Surgery, Division of Cardiothoracic Surgery, University of Alabama at Birmingham (UAB), Birmingham, AL, United States
| | - Spencer J. Melby
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University, Saint Louis, MO, United States
- Saint Louis VA Medical Center, Birmingham VA Health Care System, Birmingham, AL, United States
| | - Carlos M. Ferrario
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Louis J. Dell’Italia
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham (UAB), Birmingham, AL, United States
- Department of Veterans Affairs, Birmingham Veterans Affairs Health Care System, Birmingham, AL, United States
| |
Collapse
|
45
|
Gan PXL, Liao W, Linke KM, Mei D, Wu XD, Wong WSF. Targeting the renin angiotensin system for respiratory diseases. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 98:111-144. [PMID: 37524485 DOI: 10.1016/bs.apha.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Renin-angiotensin system (RAS) plays an indispensable role in regulating blood pressure through its effects on fluid and electrolyte balance. As an aside, cumulative evidence from experimental to clinical studies supports the notion that dysregulation of RAS contributes to the pro-inflammatory, pro-oxidative, and pro-fibrotic processes that occur in pulmonary diseases like asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and acute lung injury (ALI). Pharmacological intervention of the various RAS components can be a novel therapeutic strategy for the treatment of these respiratory diseases. In this chapter, we first give a recent update on the RAS, and then compile, review, and analyse recent reports on targeting RAS components as treatments for respiratory diseases. Inhibition of the pro-inflammatory renin, angiotensin-converting enzyme (ACE), angiotensin (Ang) II, and Ang II type 1 receptor (AT1R) axis, and activation of the protective ACE2, AT2R, Ang (1-7), and Mas receptor axis have demonstrated varying degrees of efficacies in experimental respiratory disease models or in human trials. The newly identified alamandine/Mas-related G-protein-coupled receptor member D pathway has shown some therapeutic promise as well. However, our understanding of the RAS ligand-and-receptor interactions is still inconclusive, and the modes of action and signaling cascade mediating the newly identified RAS receptors remain to be better characterized. Clinical data are obviously lacking behind the promising pre-clinical findings of certain well-established molecules targeting at different pathways of the RAS in respiratory diseases. Translational human studies should be the focus for RAS drug development in lung diseases in the next decade.
Collapse
Affiliation(s)
- Phyllis X L Gan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - W Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore; Singapore-HUJ Alliance for Research Enterprise, National University of Singapore, Singapore, Singapore
| | - Kira M Linke
- Department of Pharmacology, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - D Mei
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - X D Wu
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore; Singapore-HUJ Alliance for Research Enterprise, National University of Singapore, Singapore, Singapore; Drug Discovery and Optimization Platform, National University Health System, Singapore, Singapore.
| |
Collapse
|
46
|
Zeng C, Armando I, Yang J, Jose PA. Dopamine Receptor D 1R and D 3R and GRK4 Interaction in Hypertension. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2023; 96:95-105. [PMID: 37009199 PMCID: PMC10052590 DOI: 10.59249/mkrr9549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Essential hypertension is caused by the interaction of genetic, behavioral, and environmental factors. Abnormalities in the regulation of renal ion transport cause essential hypertension. The renal dopaminergic system, which inhibits sodium transport in all the nephron segments, is responsible for at least 50% of renal sodium excretion under conditions of moderate sodium excess. Dopaminergic signals are transduced by two families of receptors that belong to the G protein-coupled receptor (GPCR) superfamily. D1-like receptors (D1R and D5R) stimulate, while D2-like receptors (D2R, D3R, and D4R) inhibit adenylyl cyclases. The dopamine receptor subtypes, themselves, or by their interactions, regulate renal sodium transport and blood pressure. We review the role of the D1R and D3R and their interaction in the natriuresis associated with volume expansion. The D1R- and D3R-mediated inhibition of renal sodium transport involves PKA and PKC-dependent and -independent mechanisms. The D3R also increases the degradation of NHE3 via USP-mediated ubiquitinylation. Although deletion of Drd1 and Drd3 in mice causes hypertension, DRD1 polymorphisms are not always associated with human essential hypertension and polymorphisms in DRD3 are not associated with human essential hypertension. The impaired D1R and D3R function in hypertension is related to their hyper-phosphorylation; GRK4γ isoforms, R65L, A142V, and A486V, hyper-phosphorylate and desensitize D1R and D3R. The GRK4 locus is linked to and GRK4 variants are associated with high blood pressure in humans. Thus, GRK4, by itself, and by regulating genes related to the control of blood pressure may explain the "apparent" polygenic nature of essential hypertension.
Collapse
Affiliation(s)
- Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third
Military Medical University (Army Medical University), Chongqing, P. R.
China
| | - Ines Armando
- Division of Kidney Diseases and Hypertension,
Department of Medicine, The George Washington School of Medicine and Health
Sciences, Washington, DC, USA
| | - Jian Yang
- Department of Clinical Nutrition, The Third Affiliated
Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Pedro A. Jose
- Division of Kidney Diseases and Hypertension,
Department of Medicine, The George Washington School of Medicine and Health
Sciences, Washington, DC, USA
| |
Collapse
|
47
|
Heinzelman P, Romero PA. Directed evolution of angiotensin-converting enzyme 2 (ACE2) peptidase activity profiles for therapeutic applications. Protein Sci 2023; 32:e4597. [PMID: 36794431 PMCID: PMC10019445 DOI: 10.1002/pro.4597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/19/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023]
Abstract
Angiotensin-Converting Enzyme 2 (ACE2) has been investigated for its ability to beneficially modulate the Angiotensin receptor (ATR) therapeutic axis to treat multiple human diseases. Its broad substrate scope and diverse physiological roles however, limit its potential as a therapeutic agent. In this work, we address this limitation by establishing a yeast display-based liquid chromatography screen that enabled use of directed evolution to discover ACE2 variants that possess both wildtype or greater Ang-II hydrolytic activity and improved specificity toward Ang-II relative to the off-target peptide substrate Apelin-13. To obtain these results, we screened ACE2 active site libraries to reveal three substitution-tolerant positions (M360, T371 and Y510) that can be mutated to enhance ACE2's activity profile and followed up on these hits with focused double mutant libraries to further improve the enzyme. Relative to wildtype ACE2, our top variant (T371L/Y510Ile) displayed a sevenfold increase in Ang-II turnover number (kcat ), a sixfold diminished catalytic efficiency (kcat /Km ) on Apelin-13, and an overall decreased activity on other ACE2 substrates that were not directly assayed in the directed evolution screen. At physiologically relevant substrate concentrations, T371L/Y510Ile hydrolyzes as much or more Ang-II than wildtype ACE2 with concomitant Ang-II:Apelin-13 specificity improvements reaching 30-fold. Our efforts have delivered ATR axis-acting therapeutic candidates with relevance to both established and unexplored ACE2 therapeutic applications and provide a foundation for further ACE2 engineering efforts. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Pete Heinzelman
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Philip A Romero
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.,Department of Chemical & Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
48
|
Bosch A, Poglitsch M, Kannenkeril D, Kolwelter J, Striepe K, Ott C, Rauh M, Schiffer M, Achenbach S, Schmieder RE. Angiotensin pathways under therapy with empagliflozin in patients with chronic heart failure. ESC Heart Fail 2023; 10:1635-1642. [PMID: 36782339 DOI: 10.1002/ehf2.14313] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 09/05/2022] [Accepted: 01/26/2023] [Indexed: 02/15/2023] Open
Abstract
AIMS Large outcome studies demonstrated a reduction of heart failure hospitalization or cardiovascular death in patients with chronic heart failure (CHF). The renin-angiotensin system (RAS) is a key player in fluid and sodium regulation. The classic angiotensin-converting enzyme-angiotensin II-angiotensin-1 receptor axis (Ang I-ACE-Ang II receptor axis) is predominantly angiotensin II (Ang-II) induced and promotes vasoconstriction. In contrast, the angiotensin-converting-enzyme-2-angiotensin-(1-7)-Mas axis (Mas-axis) is mediated by the metabolites angiotensin-1-7 (Ang-(1-7)) and angtiotensin-1-5 (Ang-(1-5)) and exerts cardioprotective effects. METHODS We previously investigated the effect of empagliflozin on the systemic haemodynamic in patients with stable CHF (NYHA II-III) in a randomized placebo-controlled clinical trial 'Analysing the Effect of Empagliflozin on Reduction of Tissue Sodium Content in Patients With Chronic Heart Failure (ELSI)'. In a post hoc analysis, we now analysed whether empagliflozin has an effect on the RAS by measuring detailed RAS profiles (LC-MS/MS-based approach) in 72 patients from ELSI. We compared RAS parameters after 1-month and 3-months treatment with empagliflozin or placebo to baseline. The secondary goal was to analyse whether the effect of empagliflozin on RAS parameters was dependent on angiotensin-receptor-blocking (ARB) or angiotensin-converting-enzyme-inhibitor (ACEI) co-medication. RESULTS Empagliflozin medication induced a significant rise in Ang-II [68.5 pmol/L (21.3-324.2) vs. 131.5 pmol/L (34.9-564.0), P = 0.001], angiotensin-I (Ang-I) [78.7 pmol/L (21.5-236.6) vs. 125.9 pmol/L (52.6-512.9), P < 0.001], Ang-(1-7) [3.0 pmol/L (3.0-15.0) vs. 10.1 pmol/L (3.0-31.3), P = 0.006], and Ang-(1-5) [5.4 pmol/L (2.0-22.9) vs. 9.9 pmol/L (2.8-36.4), P = 0.004], which was not observed in the placebo group (baseline to 3-months treatment). A significant rise in Ang-II (206.4 pmol/L (64.2-750.6) vs. 568.2 pmol/L (164.7-1616.4), P = 0.001), Ang-(1-7) (3.0 pmol/L (3.0-14.1) vs. 15.0 pmol/L (3.0-31.3), P = 0.017), and Ang-(1-5) [12.2 pmol/L (3.8-46.6) vs. 36.4 pmol/L (11.1-90.7), P = 0.001] under empagliflozin treatment was only seen in the subgroup of patients with ARB co-medication, whereas no change of Ang-II (16.7 pmol/L (2.0-60.8) vs. 26.4 pmol/L (10.7-63.4), P = 0.469), Ang-(1-7) (6.6 pmol/L (3.0-20.7) vs. 10.5 pmol/L (3.0-50.5), P = 0.221), and Ang-(1-5) (2.7 pmol/L (2.0-8.4) vs. 2.8 pmol/L (2.0-6.9), P = 0.851) was observed in patients with empagliflozin that were on ACEI co-medication (baseline to 3-months treatment). CONCLUSIONS Our data indicate that empagliflozin might lead to an activation of both the Ang I-ACE-Ang II receptor axis and the Mas-axis pathway. Activation of the Ang I-ACE-Ang II receptor axis and the protective Mas-axis pathway after initiating treatment with empagliflozin was only seen in patients with ARB co-medication, in contrast to co-medication with ACEI.
Collapse
Affiliation(s)
- Agnes Bosch
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | - Dennis Kannenkeril
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Julie Kolwelter
- Department of Cardiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Kristina Striepe
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christian Ott
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Department of Nephrology and Hypertension, Paracelsus Medical University, Nürnberg, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stephan Achenbach
- Department of Cardiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Roland E Schmieder
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
49
|
Gong X, Khan A, Wani MY, Ahmad A, Duse A. COVID-19: A state of art on immunological responses, mutations, and treatment modalities in riposte. J Infect Public Health 2023; 16:233-249. [PMID: 36603376 PMCID: PMC9798670 DOI: 10.1016/j.jiph.2022.12.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/25/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Over the last few years, the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) unleashed a global public health catastrophe that had a substantial influence on human physical and mental health, the global economy, and socio-political dynamics. SARS-CoV-2 is a respiratory pathogen and the cause of ongoing COVID-19 pandemic, which testified how unprepared humans are for pandemics. Scientists and policymakers continue to face challenges in developing ideal therapeutic agents and vaccines, while at the same time deciphering the pathology and immunology of SARS-CoV-2. Challenges in the early part of the pandemic included the rapid development of diagnostic assays, vaccines, and therapeutic agents. The ongoing transmission of COVID-19 is coupled with the emergence of viral variants that differ in their transmission efficiency, virulence, and vaccine susceptibility, thus complicating the spread of the pandemic. Our understanding of how the human immune system responds to these viruses as well as the patient groups (such as the elderly and immunocompromised individuals) who are often more susceptible to serious illness have both been aided by this epidemic. COVID-19 causes different symptoms to occur at different stages of infection, making it difficult to determine distinct treatment regimens employed for the various clinical phases of the disease. Unsurprisingly, determining the efficacy of currently available medications and developing novel therapeutic strategies have been a process of trial and error. The global scientific community collaborated to research and develop vaccines at a neck-breaking speed. This review summarises the overall picture of the COVID-19 pandemic, different mutations in SARS-CoV-2, immune response, and the treatment modalities against SARS-CoV-2.
Collapse
Affiliation(s)
- Xiaolong Gong
- Department of Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Amber Khan
- Department of Clinical Haematology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mohmmad Younus Wani
- Department of Chemistry, College of Science, University of Jeddah, P.O. Box 80327, Jeddah 21589, Kingdom of Saudi Arabia
| | - Aijaz Ahmad
- Department of Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa,Division of Infection Control, Charlotte Maxeke Johannesburg Academic Hospital, National Health Laboratory Service, Johannesburg, South Africa,Corresponding author at: Department of Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Adriano Duse
- Department of Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa,Division of Infection Control, Charlotte Maxeke Johannesburg Academic Hospital, National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
50
|
Karasaki K, Kokubo H, Bumdelger B, Kaji N, Sakai C, Ishida M, Yoshizumi M. Angiotensin II Type 1 Receptor Blocker Prevents Abdominal Aortic Aneurysm Progression in Osteoprotegerin-Deficient Mice via Upregulation of Angiotensin (1-7). J Am Heart Assoc 2023; 12:e027589. [PMID: 36718875 PMCID: PMC9973615 DOI: 10.1161/jaha.122.027589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Background Angiotensin II type 1 receptor blockers (ARBs) have been shown to limit the growth of abdominal aortic aneurysm (AAA), but their efficacy is controversial. This study aimed to investigate the molecular mechanism underlying the protective effect of ARBs against AAA progression. Methods and Results Olmesartan, an ARB, was administered to wild-type and osteoprotegerin-knockout (Opg-KO) mice starting 2 weeks before direct application of CaCl2 to aortas to induce AAA. The protective effect of olmesartan against AAA in wild-type and Opg-KO mice was compared at 6 weeks after AAA induction. Olmesartan prevented AAA progression in Opg-KO mice, including excessive aortic dilatation and collapse of tunica media, but not in wild-type mice. Deficiency of the Opg gene is known to cause excessive activation of the tumor necrosis factor-related apoptosis-inducing ligand-induced c-Jun N-terminal kinase/matrix metalloproteinase 9 pathway, resulting in prolonged AAA progression. Olmesartan attenuated the upregulation of phosphorylated c-Jun N-terminal kinase and matrix metalloproteinase 9 expression in the aortic wall of Opg-KO mice. In cultured vascular smooth muscle cells, tumor necrosis factor-related apoptosis-inducing ligand-induced c-Jun N-terminal kinase phosphorylation and matrix metalloproteinase 9 expression were inhibited by angiotensin (1-7), the circulating levels of which are increased by ARBs. Furthermore, administering an angiotensin (1-7) antagonist to Opg-KO mice diminished the protective effect of olmesartan against AAA progression. Conclusions Olmesartan prevented AAA progression in Opg-KO mice by upregulating angiotensin (1-7), suggesting that angiotensin (1-7) may be a key factor that mediates the protective effect of ARBs.
Collapse
Affiliation(s)
- Kohei Karasaki
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Hiroki Kokubo
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Batmunkh Bumdelger
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Nobuchika Kaji
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Chiemi Sakai
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Mari Ishida
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Masao Yoshizumi
- Department of Cardiovascular Physiology and Medicine, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| |
Collapse
|