1
|
Dong XQ, Zhang YH, Luo J, Li MJ, Ma LQ, Qi YT, Miao YL. Keratin 1 modulates intestinal barrier and immune response via kallikrein kinin system in ulcerative colitis. World J Gastroenterol 2025; 31:102070. [PMID: 39958441 PMCID: PMC11752705 DOI: 10.3748/wjg.v31.i6.102070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/06/2024] [Accepted: 12/19/2024] [Indexed: 01/10/2025] Open
Abstract
BACKGROUND External factors in ulcerative colitis (UC) exacerbate colonic epithelial permeability and inflammatory responses. Keratin 1 (KRT1) is crucial in regulating these alterations, but its specific role in the progression of UC remains to be fully elucidated. AIM To explore the role and mechanisms of KRT1 in the regulation of colonic epithelial permeability and inflammation in UC. METHODS A KRT1 antibody concentration gradient test, along with a dextran sulfate sodium (DSS)-induced animal model, was implemented to investigate the role of KRT1 in modulating the activation of the kallikrein kinin system (KKS) and the cleavage of bradykinin (BK)/high molecular weight kininogen (HK) in UC. RESULTS Treatment with KRT1 antibody in Caco-2 cells suppressed cell proliferation, induced apoptosis, reduced HK expression, and increased BK expression. It further downregulated intestinal barrier proteins, including occludin, zonula occludens-1, and claudin, and negatively impacted the coagulation factor XII. These changes led to enhanced activation of BK and HK cleavage, thereby intensifying KKS-mediated inflammation in UC. In the DSS-induced mouse model, administration of KRT1 antibody mitigated colonic injury, increased colon length, alleviated weight loss, and suppressed inflammatory cytokines such as interleukin (IL)-1, IL-6, tumor necrosis factor-α. It also facilitated repair of the intestinal barrier, reducing DSS-induced injury. CONCLUSION KRT1 inhibits BK expression, suppresses inflammatory cytokines, and enhances markers of intestinal barrier function, thus ameliorating colonic damage and maintaining barrier integrity. KRT1 is a viable therapeutic target for UC.
Collapse
Affiliation(s)
- Xiang-Qian Dong
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan Province, China
| | - Ying-Hui Zhang
- Department of Gastroenterology, Affiliated Hospital of Yunnan University, Kunming 650021, Yunnan Province, China
| | - Juan Luo
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan Province, China
| | - Mao-Juan Li
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan Province, China
| | - Lan-Qing Ma
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan Province, China
| | - Ya-Ting Qi
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan Province, China
| | - Ying-Lei Miao
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Diseases, Kunming 650032, Yunnan Province, China
| |
Collapse
|
2
|
Zhang Q, Ruan H, Wang X, Luo A, Ran X. Ulinastatin attenuated cardiac ischaemia/reperfusion injury by suppressing activation of the tissue kallikrein-kinin system. Br J Pharmacol 2024; 181:4988-5008. [PMID: 39294926 DOI: 10.1111/bph.16477] [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: 08/14/2023] [Revised: 02/29/2024] [Accepted: 05/13/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Ulinastatin has beneficial effects in patients undergoing coronary artery bypass grafting (CABG) surgery due to its anti-inflammatory properties, but the underlying mechanism remains unclear. EXPERIMENTAL APPROACH We used samples from patients undergoing CABG, a model of cardiac ischaemia-reperfusion injury (IRI) in mice and murine cardiac endothelial cell cultures to investigate links between ulinastatin, the kallikrein-kinin system (KKS), endothelial dysfunction and cardiac inflammation in the response to ischaemia/reperfusion injury (IRI). These links were assessed using clinical investigations, in vitro and in vivo experiments and RNA sequencing analysis. KEY RESULTS Ulinastatin inhibited the activity of tissue kallikrein, a key enzyme of the KKS, at 24 h after CABG surgery, which was verified in our murine cardiac ischaemia-reperfusion model. Under normal conditions, ulinastatin only inhibited kallikrein activity but did not affect bradykinin (B1/B2) receptors. Ulinastatin protected against IRI, in vivo and in vitro, by suppressing activation of the kallikrein-kinin system and down-regulating B1/B2 receptor-related signalling pathways including ERK/ iNOS, which resulted in enhanced endothelial barrier function, mitigation of inflammation and oedema, decreased infarct size, improved cardiac function and decreased mortality. Inhibition of kallikrein and knockdown of B1, but not B2 receptors prevented ERK translocation into the nucleus, reducing reperfusion-induced injury in murine cardiac endothelial cells. CONCLUSIONS AND IMPLICATIONS Treatment with ulinastatin exerts a protective influence on cardiac reperfusion by suppressing activation of the kallikrein-kinin system. Our findings highlight the potential of targeting kallikrein /bradykinin receptors to alleviate endothelial dysfunction, thus improving cardiac IRI.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hang Ruan
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ailin Luo
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Ran
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Kamollerd C, Uopasai S, Kamollerd T, Lapyuneyong N, Taoto C, Iamsaard S, Tangsrisakda N. Associations between androgen receptor and tyrosine phosphorylated protein expressions in rat prostate gland. BRAZ J BIOL 2024; 84:e285484. [PMID: 39607256 DOI: 10.1590/1519-6984.285484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/19/2024] [Indexed: 11/29/2024] Open
Abstract
Mammalian prostate gland plays a role in alkaline substance synthesis including proteins. These functions are depending on glandular maturation and testosterone-androgen receptor (AR) dependent actions. Since tyrosine phosphorylated (TyrPho) proteins, also important for secreting pathways, have been localized in the androgen dependent organs, association between AR and TyrPho protein expressions in prostate is still unknown. This study aimed to investigate the changes of such proteins in prostate gland of male castrated rats. Nine prepubertal and adult twenty-two adult male rats were divided into the prepubertal (Pre, n=9), Sham (n=6), castrate for 3 (Cas-3, n=8) and for 7 (Cas-7, n=8) days groups, respectively. Serum testosterone level was determined. Histology and AR localization in each prostatic lobe were observed. TyrPho and AR protein expressions were also examined. The results showed undetectable testosterone level and low AR expression in Pre and Cas prostates with the decreased size. Few histopathologies were found in Cas groups. In ventral lobe, a Tyrpho protein was increased at the 48 kDa but the 52, 33, and 26 kDas were decreased in the Pre and Cas groups. For dorsolateral lobe, they were decreased at 33 and 30 kDas in Pre group and only 30 kDa was decreased in Cas-3 group. In the anterior lobe, the TyrPho proteins 57, 49, 39, 30, and 26 kDas were decreased in Pre group while 57, 30, and 26 kDas were decreased in Cas-3 group. In conclusion, the alterations of testosterone level and AR expressions associate with TyrPho protein expressions in prostate gland during development.
Collapse
Affiliation(s)
- C Kamollerd
- Khon Kaen University, Faculty of Veterinary Medicine, Department of Anatomy, Khon Kaen, Thailand
| | - S Uopasai
- Khon Kaen University, Faculty of Veterinary Medicine, Department of Anatomy, Khon Kaen, Thailand
| | - T Kamollerd
- Khon Kaen University, Faculty of Medicine, Department of Anatomy, Khon Kaen, Thailand
| | - N Lapyuneyong
- Khon Kaen University, Faculty of Medicine, Department of Anatomy, Khon Kaen, Thailand
| | - C Taoto
- Khon Kaen University, Faculty of Medicine, Department of Anatomy, Khon Kaen, Thailand
| | - S Iamsaard
- Khon Kaen University, Faculty of Medicine, Department of Anatomy, Khon Kaen, Thailand
| | - N Tangsrisakda
- Khon Kaen University, Faculty of Medicine, Department of Anatomy, Khon Kaen, Thailand
| |
Collapse
|
4
|
Liu XH, Liu XT, Wu Y, Li SA, Ren KD, Cheng M, Huang B, Yang Y, Liu PP. Broadening Horizons: Exploring the Cathepsin Family as Therapeutic Targets for Alzheimer's Disease. Aging Dis 2024:AD.2024.0456. [PMID: 39122455 DOI: 10.14336/ad.2024.0456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/02/2024] [Indexed: 08/12/2024] Open
Abstract
Alzheimer's disease (AD) is an intricate neurodegenerative disorder characterized by the accumulation of misfolded proteins, including beta-amyloid (Aβ) and tau, leading to cognitive decline. Despite decades of research, the precise mechanisms underlying its onset and progression remain elusive. Cathepsins are a family of lysosomal enzymes that play vital roles in cellular processes, including protein degradation and regulation of immune responses. Emerging evidence suggests that cathepsins may be involved in AD pathogenesis. Cathepsins can influence the activation of microglia and astrocytes, the resident immune cells in the brain. However, cathepsin dysfunction may lead to the accumulation of misfolded proteins, notably Aβ and tau. In addition, dysregulated cathepsin activity may induce an exaggerated immune response, promoting chronic inflammation and neuronal dysfunction in patients with AD. By unraveling the classification, functions, and roles of cathepsins in AD's pathogenesis, this review sheds light on their intricate involvement in this devastating disease. Targeting cathepsin activity could be a promising and novel approach for mitigating the pathological processes that contribute to AD, providing new avenues for its treatment and prevention.
Collapse
Affiliation(s)
- Xiao-Hui Liu
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiao-Tong Liu
- Clinical Laboratory, the First Hospital of Yongnian District, Yongnian, Hebei, China
| | - Yue Wu
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shu-Ang Li
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kai-Di Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Meng Cheng
- Translational Medical Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bing Huang
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, China
| | - Yang Yang
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pei-Pei Liu
- Clinical Systems Biology Laboratories, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Padín JF, Pérez-Ortiz JM, Redondo-Calvo FJ. Aprotinin (I): Understanding the Role of Host Proteases in COVID-19 and the Importance of Pharmacologically Regulating Their Function. Int J Mol Sci 2024; 25:7553. [PMID: 39062796 PMCID: PMC11277036 DOI: 10.3390/ijms25147553] [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: 05/27/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Proteases are produced and released in the mucosal cells of the respiratory tract and have important physiological functions, for example, maintaining airway humidification to allow proper gas exchange. The infectious mechanism of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), takes advantage of host proteases in two ways: to change the spatial conformation of the spike (S) protein via endoproteolysis (e.g., transmembrane serine protease type 2 (TMPRSS2)) and as a target to anchor to epithelial cells (e.g., angiotensin-converting enzyme 2 (ACE2)). This infectious process leads to an imbalance in the mucosa between the release and action of proteases versus regulation by anti-proteases, which contributes to the exacerbation of the inflammatory and prothrombotic response in COVID-19. In this article, we describe the most important proteases that are affected in COVID-19, and how their overactivation affects the three main physiological systems in which they participate: the complement system and the kinin-kallikrein system (KKS), which both form part of the contact system of innate immunity, and the renin-angiotensin-aldosterone system (RAAS). We aim to elucidate the pathophysiological bases of COVID-19 in the context of the imbalance between the action of proteases and anti-proteases to understand the mechanism of aprotinin action (a panprotease inhibitor). In a second-part review, titled "Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions", we explain in depth the pharmacodynamics, pharmacokinetics, toxicity, and use of aprotinin as an antiviral drug.
Collapse
Affiliation(s)
- Juan Fernando Padín
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain;
| | - José Manuel Pérez-Ortiz
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Francisco Javier Redondo-Calvo
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain;
- Department of Anaesthesiology and Critical Care Medicine, University General Hospital, 13005 Ciudad Real, Spain
- Translational Research Unit, University General Hospital and Research Institute of Castilla-La Mancha (IDISCAM), 13005 Ciudad Real, Spain
| |
Collapse
|
6
|
Padín JF, Pérez-Ortiz JM, Redondo-Calvo FJ. Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions. Int J Mol Sci 2024; 25:7209. [PMID: 39000315 PMCID: PMC11241800 DOI: 10.3390/ijms25137209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Aprotinin is a broad-spectrum inhibitor of human proteases that has been approved for the treatment of bleeding in single coronary artery bypass surgery because of its potent antifibrinolytic actions. Following the outbreak of the COVID-19 pandemic, there was an urgent need to find new antiviral drugs. Aprotinin is a good candidate for therapeutic repositioning as a broad-spectrum antiviral drug and for treating the symptomatic processes that characterise viral respiratory diseases, including COVID-19. This is due to its strong pharmacological ability to inhibit a plethora of host proteases used by respiratory viruses in their infective mechanisms. The proteases allow the cleavage and conformational change of proteins that make up their viral capsid, and thus enable them to anchor themselves by recognition of their target in the epithelial cell. In addition, the activation of these proteases initiates the inflammatory process that triggers the infection. The attraction of the drug is not only its pharmacodynamic characteristics but also the possibility of administration by the inhalation route, avoiding unwanted systemic effects. This, together with the low cost of treatment (≈2 Euro/dose), makes it a good candidate to reach countries with lower economic means. In this article, we will discuss the pharmacodynamic, pharmacokinetic, and toxicological characteristics of aprotinin administered by the inhalation route; analyse the main advances in our knowledge of this medication; and the future directions that should be taken in research in order to reposition this medication in therapeutics.
Collapse
Affiliation(s)
- Juan-Fernando Padín
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain
| | - José Manuel Pérez-Ortiz
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Francisco Javier Redondo-Calvo
- Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain
- Department of Anaesthesiology and Critical Care Medicine, University General Hospital, 13005 Ciudad Real, Spain
- Translational Research Unit, University General Hospital and Research Institute of Castilla-La Mancha (IDISCAM), 13005 Ciudad Real, Spain
| |
Collapse
|
7
|
Wisniewski P, Gangnus T, Burckhardt BB. Recent advances in the discovery and development of drugs targeting the kallikrein-kinin system. J Transl Med 2024; 22:388. [PMID: 38671481 PMCID: PMC11046790 DOI: 10.1186/s12967-024-05216-5] [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: 01/12/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The kallikrein-kinin system is a key regulatory cascade involved in blood pressure maintenance, hemostasis, inflammation and renal function. Currently, approved drugs remain limited to the rare disease hereditary angioedema. However, growing interest in this system is indicated by an increasing number of promising drug candidates for further indications. METHODS To provide an overview of current drug development, a two-stage literature search was conducted between March and December 2023 to identify drug candidates with targets in the kallikrein-kinin system. First, drug candidates were identified using PubMed and Clinicaltrials.gov. Second, the latest publications/results for these compounds were searched in PubMed, Clinicaltrials.gov and Google Scholar. The findings were categorized by target, stage of development, and intended indication. RESULTS The search identified 68 drugs, of which 10 are approved, 25 are in clinical development, and 33 in preclinical development. The three most studied indications included diabetic retinopathy, thromboprophylaxis and hereditary angioedema. The latter is still an indication for most of the drug candidates close to regulatory approval (3 out of 4). For the emerging indications, promising new drug candidates in clinical development are ixodes ricinus-contact phase inhibitor for thromboprophylaxis and RZ402 and THR-149 for the treatment of diabetic macular edema (all phase 2). CONCLUSION The therapeutic impact of targeting the kallikrein-kinin system is no longer limited to the treatment of hereditary angioedema. Ongoing research on other diseases demonstrates the potential of therapeutic interventions targeting the kallikrein-kinin system and will provide further treatment options for patients in the future.
Collapse
Affiliation(s)
- Petra Wisniewski
- Individualized Pharmacotherapy, Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, 48149, Münster, Germany
| | - Tanja Gangnus
- Individualized Pharmacotherapy, Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, 48149, Münster, Germany
| | - Bjoern B Burckhardt
- Individualized Pharmacotherapy, Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, 48149, Münster, Germany.
| |
Collapse
|
8
|
Kaschina E, Lauer D, Lange C, Unger T. Angiotensin AT 2 receptors reduce inflammation and fibrosis in cardiovascular remodeling. Biochem Pharmacol 2024; 222:116062. [PMID: 38369211 DOI: 10.1016/j.bcp.2024.116062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/04/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
The angiotensin AT2 receptor (AT2R), an important member of the "protective arm" of the renin-angiotensin system (RAS), has been recently defined as a therapeutic target in different pathological conditions. The AT2R activates complex signalling pathways linked to cellular proliferation, differentiation, anti-inflammation, antifibrosis, and induction or inhibition of apoptosis. The anti-inflammatory effect of AT2R activation is commonly associated with reduced fibrosis in different models. Current discoveries demonstrated a direct impact of AT2Rs on the regulation of cytokines, transforming growth factor beta1 (TGF-beta1), matrix metalloproteases (MMPs), and synthesis of the extracellular matrix components. This review article summarizes current knowledge on the AT2R in regard to immunity, inflammation and fibrosis in the heart and blood vessels. In particular, the differential influence of the AT2R on cardiovascular remodeling in preclinical models of myocardial infarction, heart failure and aneurysm formation are discussed. Overall, these studies demonstrate that AT2R stimulation represents a promising therapeutic approach to counteract myocardial and aortic damage in cardiovascular diseases.
Collapse
Affiliation(s)
- Elena Kaschina
- Charité - Universitätsmedizin Berlin, Institute of Pharmacology, Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Berlin, Germany.
| | - Dilyara Lauer
- Charité - Universitätsmedizin Berlin, Institute of Pharmacology, Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Berlin, Germany
| | - Christoph Lange
- Charité - Universitätsmedizin Berlin, Institute of Pharmacology, Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Berlin, Germany
| | - Thomas Unger
- CARIM - School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
9
|
Aly R, Dogan YE, Bala N, Lugo C, Darwish S, Shoemaker L, Alli AA. Dysregulation of kidney proteases in the pathogenesis of hypertension following unilateral nephrectomy in juvenile mice. Am J Transl Res 2024; 16:544-556. [PMID: 38463588 PMCID: PMC10918144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/12/2023] [Indexed: 03/12/2024]
Abstract
BACKGROUND Unliteral nephrectomy (UNX) results in the reduction of kidney mass. The remaining kidney undergoes compensatory renal growth via hypertrophy of the glomeruli and renal tubules to maintain a normal glomerular filtration rate (GFR). These compensatory mechanisms result in increased capillary pressure and glomerular hyperfiltration to increase single nephron GFR. Over time, hyperfiltration may lead to kidney scarring and the development of hypertension. OBJECTIVES The first objective of this study was to test the hypothesis that a 50% reduction in functioning nephrons in juvenile mice leads to increased blood pressure over a 24-hour phase. The second objective was to test the hypothesis that UNX leads to changes in the expression and activity of kidney proteases in juvenile mice. METHODS Eight male C57B6 juvenile wild-type mice were subject to UNX and an equal number of mice were subject to sham (SH) surgery. Metabolic cage studies were performed for 5 weeks to collect urine produced during the inactive and active phases. Blood pressure was measured using the tail cuff method twice weekly and tail blood was collected on different days during the inactive or active phase of each animal. The mice were euthanized at the age of 9 weeks. Western blotting and immunohistochemistry were performed to investigate changes in renal protein expression of various cathepsins and renal kallikrein 1 (KLK1) between the two groups. Protease activity assays were performed using kidney lysates and urine samples from each group. RESULTS Compared to the SH group, UNX mice showed a persistent increase in blood pressure at week 3 which progressed toward the end of the study at week 5 of age. Cathepsin B, D, and S expression and activity were up-regulated in kidney cortex lysates from UNX mice compared to the SH control group. KLK1 protein expression was down-regulated and urinary nitric oxide excretion was decreased in UNX mice compared to the SH control group. CONCLUSION UNX results in the development of persistent and progressive hypertension. Down-regulation of KLK1 and up-regulation of various cathepsins may contribute to the development of hypertension via multiple mechanisms including a decrease in nitric oxide (NO) production.
Collapse
Affiliation(s)
- Rasha Aly
- Department of Pediatrics, Division of Pediatric Nephrology, University of Florida College of MedicineGainesville, FL, USA
| | - Yunus E Dogan
- Department of Physiology and Aging, University of Florida College of MedicineGainesville, FL, USA
- Department of Pediatrics, Faculty of Medicine, Erciyes UniversityKayseri, Turkey
| | - Niharika Bala
- Department of Physiology and Aging, University of Florida College of MedicineGainesville, FL, USA
| | - Carlos Lugo
- Department of Physiology and Aging, University of Florida College of MedicineGainesville, FL, USA
| | - Seena Darwish
- Department of Physiology and Aging, University of Florida College of MedicineGainesville, FL, USA
| | - Lawrence Shoemaker
- Department of Pediatrics, Division of Pediatric Nephrology, University of Florida College of MedicineGainesville, FL, USA
| | - Abdel A Alli
- Department of Pediatrics, Division of Pediatric Nephrology, University of Florida College of MedicineGainesville, FL, USA
- Department of Physiology and Aging, University of Florida College of MedicineGainesville, FL, USA
- Department of Medicine, Division of Nephrology, Hypertension and Renal Transplantation, University of Florida College of MedicineGainesville, FL, USA
| |
Collapse
|
10
|
Ibrahim Z, Khan NA, Qaisar R, Saleh MA, Siddiqui R, Al-Hroub HM, Giddey AD, Semreen MH, Soares NC, Elmoselhi AB. Serum multi-omics analysis in hindlimb unloading mice model: Insights into systemic molecular changes and potential diagnostic and therapeutic biomarkers. Heliyon 2024; 10:e23592. [PMID: 38187258 PMCID: PMC10770503 DOI: 10.1016/j.heliyon.2023.e23592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/09/2024] Open
Abstract
Microgravity, in space travel and prolonged bed rest conditions, induces cardiovascular deconditioning along with skeletal muscle mass loss and weakness. The findings of microgravity research may also aid in the understanding and treatment of human health conditions on Earth such as muscle atrophy, and cardiovascular diseases. Due to the paucity of biomarkers and the unknown underlying mechanisms of cardiovascular and skeletal muscle deconditioning in these environments, there are insufficient diagnostic and preventative measures. In this study, we employed hindlimb unloading (HU) mouse model, which mimics astronauts in space and bedridden patients, to first evaluate cardiovascular and skeletal muscle function, followed by proteomics and metabolomics LC-MS/MS-based analysis using serum samples. Three weeks of unloading caused changes in the function of the cardiovascular system in c57/Bl6 mice, as seen by a decrease in mean arterial pressure and heart weight. Unloading for three weeks also changed skeletal muscle function, causing a loss in grip strength in HU mice and atrophy of skeletal muscle indicated by a reduction in muscle mass. These modifications were partially reversed by a two-week recovery period of reloading condition, emphasizing the significance of the recovery process. Proteomics analysis revealed 12 dysregulated proteins among the groups, such as phospholipid transfer protein, Carbonic anhydrase 3, Parvalbumin alpha, Major urinary protein 20 (Mup20), Thrombospondin-1, and Apolipoprotein C-IV. On the other hand, metabolomics analysis showed altered metabolites among the groups such as inosine, hypoxanthine, xanthosine, sphinganine, l-valine, 3,4-Dihydroxyphenylglycol, and l-Glutamic acid. The joint data analysis revealed that HU conditions mainly impacted pathways such as ABC transporters, complement and coagulation cascades, nitrogen metabolism, and purine metabolism. Overall, our results indicate that microgravity environment induces significant alterations in the function, proteins, and metabolites of these mice. These observations suggest the potential utilization of these proteins and metabolites as novel biomarkers for assessing and mitigating cardiovascular and skeletal muscle deconditioning associated with such conditions.
Collapse
Affiliation(s)
- Zeinab Ibrahim
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Naveed A. Khan
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Rizwan Qaisar
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohamed A. Saleh
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ruqaiyyah Siddiqui
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University Edinburgh, EH14 4AS UK
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Hamza M. Al-Hroub
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Alexander D. Giddey
- Center for Applied and Translational Genomics, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Mohammad Harb Semreen
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Nelson C. Soares
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Av. Padre Cruz, Lisbon, 1649-016, Portugal
- Centre for Toxicogenomics and Human Health (ToxOmics), NOVA School/ Faculdade de Lisboa, Lisbon, Portugal
| | - Adel B. Elmoselhi
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
11
|
Aufy M, Hussein AM, Stojanovic T, Studenik CR, Kotob MH. Proteolytic Activation of the Epithelial Sodium Channel (ENaC): Its Mechanisms and Implications. Int J Mol Sci 2023; 24:17563. [PMID: 38139392 PMCID: PMC10743461 DOI: 10.3390/ijms242417563] [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: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Epithelial sodium channel (ENaC) are integral to maintaining salt and water homeostasis in various biological tissues, including the kidney, lung, and colon. They enable the selective reabsorption of sodium ions, which is a process critical for controlling blood pressure, electrolyte balance, and overall fluid volume. ENaC activity is finely controlled through proteolytic activation, a process wherein specific enzymes, or proteases, cleave ENaC subunits, resulting in channel activation and increased sodium reabsorption. This regulatory mechanism plays a pivotal role in adapting sodium transport to different physiological conditions. In this review article, we provide an in-depth exploration of the role of proteolytic activation in regulating ENaC activity. We elucidate the involvement of various proteases, including furin-like convertases, cysteine, and serine proteases, and detail the precise cleavage sites and regulatory mechanisms underlying ENaC activation by these proteases. We also discuss the physiological implications of proteolytic ENaC activation, focusing on its involvement in blood pressure regulation, pulmonary function, and intestinal sodium absorption. Understanding the mechanisms and consequences of ENaC proteolytic activation provides valuable insights into the pathophysiology of various diseases, including hypertension, pulmonary disorders, and various gastrointestinal conditions. Moreover, we discuss the potential therapeutic avenues that emerge from understanding these mechanisms, offering new possibilities for managing diseases associated with ENaC dysfunction. In summary, this review provides a comprehensive discussion of the intricate interplay between proteases and ENaC, emphasizing the significance of proteolytic activation in maintaining sodium and fluid balance in both health and disease.
Collapse
Affiliation(s)
- Mohammed Aufy
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
| | - Ahmed M. Hussein
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
- Department of Zoology, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt
| | - Tamara Stojanovic
- Programme for Proteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Christian R. Studenik
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
| | - Mohamed H. Kotob
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
- Department of Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut 71515, Egypt
| |
Collapse
|
12
|
Richman TR, Ermer JA, Baker J, Siira SJ, Kile BT, Linden MD, Rackham O, Filipovska A. Mitochondrial gene expression is required for platelet function and blood clotting. Cell Rep 2023; 42:113312. [PMID: 37889747 DOI: 10.1016/j.celrep.2023.113312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/20/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Platelets are anucleate blood cells that contain mitochondria and regulate blood clotting in response to injury. Mitochondria contain their own gene expression machinery that relies on nuclear-encoded factors for the biogenesis of the oxidative phosphorylation system to produce energy required for thrombosis. The autonomy of the mitochondrial gene expression machinery from the nucleus is unclear, and platelets provide a valuable model to understand its importance in anucleate cells. Here, we conditionally delete Elac2, Ptcd1, or Mtif3 in platelets, which are essential for mitochondrial gene expression at the level of RNA processing, stability, or translation, respectively. Loss of ELAC2, PTCD1, or MTIF3 leads to increased megakaryocyte ploidy, elevated circulating levels of reticulated platelets, thrombocytopenia, and consequent extended bleeding time. Impaired mitochondrial gene expression reduces agonist-induced platelet activation. Transcriptomic and proteomic analyses show that mitochondrial gene expression is required for fibrinolysis, hemostasis, and blood coagulation in response to injury.
Collapse
Affiliation(s)
- Tara R Richman
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia; ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA 6009, Australia; Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, WA 6009, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, WA, Australia
| | - Judith A Ermer
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia; ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA 6009, Australia; Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - Jessica Baker
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia; ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA 6009, Australia; Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, WA 6009, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, WA, Australia
| | - Stefan J Siira
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia; ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA 6009, Australia; Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, WA 6009, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, WA, Australia
| | - Benjamin T Kile
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Matthew D Linden
- Pathology and Laboratory Science, The University of Western Australia, Perth, WA, Australia
| | - Oliver Rackham
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia; ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA 6009, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, WA, Australia; Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
| | - Aleksandra Filipovska
- ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA 6009, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, WA, Australia.
| |
Collapse
|
13
|
Lu L, Li DX, Chen W, Li GS, Hao P. Bradykinin-(1-9) mitigates autophagy through upregulating PI3K/Akt in rats with myocardial infarction. Biochem Biophys Res Commun 2023; 660:35-42. [PMID: 37060829 DOI: 10.1016/j.bbrc.2023.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/26/2023] [Accepted: 04/04/2023] [Indexed: 04/17/2023]
Abstract
The cardioprotective mechanisms of bradykinin-(1-9) in myocardial infarction were unclear. We investigated the effect of bradykinin-(1-9) on cardiac function, fibrosis, and autophagy induced by myocardial infarction and identified the mechanisms involved. To investigate the cardioprotective effect of bradykinin-(1-9), various doses of bradykinin-(1-9), its B2 receptor blocker HOE140, or their combination were administered to rats via subcutaneous osmotic minipump implantation before myocardial infarction. After 2 days, myocardial infarction was induced by ligation of the left anterior descending coronary artery. After 2 weeks, echocardiographic measurements and euthanasia were performed. Bradykinin-(1-9) treatment attenuated left ventricular dysfunction, fibrosis, and autophagy in rats with myocardial infarction, which was partially reversed by HOE140 administration. Moreover, the downregulatory effect of bradykinin-(1-9) on autophagy was partially reversed by combination with the PI3K inhibitor LY294002. Thus, bradykinin-(1-9) inhibits myocardial infarction-induced cardiomyocyte autophagy by upregulating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Lin Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China; Department of Cardiovascular Medicine, the Third Affiliated Hospital of Shandong First Medical University, Jinan, 250031, Shandong Province, China
| | - Dai-Xu Li
- Department of Cardiovascular Medicine, The Fourth People's Hospital of Jinan, Jinan, 250031, Shandong Province, China
| | - Wei Chen
- Department of Cardiovascular Medicine, the Third Affiliated Hospital of Shandong First Medical University, Jinan, 250031, Shandong Province, China
| | - Gui-Shuang Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China.
| | - Panpan Hao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
14
|
Shen JK, Zhang HT. Function and structure of bradykinin receptor 2 for drug discovery. Acta Pharmacol Sin 2023; 44:489-498. [PMID: 36075965 PMCID: PMC9453710 DOI: 10.1038/s41401-022-00982-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022]
Abstract
Type 2 bradykinin receptor (B2R) is an essential G protein-coupled receptor (GPCR) that regulates the cardiovascular system as a vasodepressor. Dysfunction of B2R is also closely related to cancers and hereditary angioedema (HAE). Although several B2R agonists and antagonists have been developed, icatibant is the only B2R antagonist clinically used for treating HAE. The recently determined structures of B2R have provided molecular insights into the functions and regulation of B2R, which shed light on structure-based drug design for the treatment of B2R-related diseases. In this review, we summarize the structure and function of B2R in relation to drug discovery and discuss future research directions to elucidate the remaining unknown functions of B2R dimerization.
Collapse
Affiliation(s)
- Jin-Kang Shen
- Hangzhou Institute of Innovative Medicine, Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hai-Tao Zhang
- Hangzhou Institute of Innovative Medicine, Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
15
|
Dietary Mg Supplementation Decreases Oxidative Stress, Inflammation, and Vascular Dysfunction in an Experimental Model of Metabolic Syndrome with Renal Failure. Antioxidants (Basel) 2023; 12:antiox12020283. [PMID: 36829843 PMCID: PMC9952257 DOI: 10.3390/antiox12020283] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) and chronic kidney disease (CKD) are commonly associated with cardiovascular disease (CVD) and in these patients Mg concentration is usually decreased. This study evaluated whether a dietary Mg supplementation might attenuate vascular dysfunction through the modulation of oxidative stress and inflammation in concurrent MetS and CKD. METHODS A rat model of MetS (Zucker strain) with CKD (5/6 nephrectomy, Nx) was used. Nephrectomized animals were fed a normal 0.1%Mg (MetS+Nx+Mg0.1%) or a supplemented 0.6%Mg (MetS+Nx+Mg0.6%) diet; Sham-operated rats with MetS receiving 0.1%Mg were used as controls. RESULTS As compared to controls, the MetS+Nx-Mg0.1% group showed a significant increase in oxidative stress and inflammation biomarkers (lipid peroxidation and aortic interleukin-1b and -6 expression) and Endothelin-1 levels, a decrease in nitric oxide and a worsening in uremia and MetS associated pathology as hypertension, and abnormal glucose and lipid profile. Moreover, proteomic evaluation revealed changes mainly related to lipid metabolism and CVD markers. By contrast, in the MetS+Nx+Mg0.6% group, these parameters remained largely similar to controls. CONCLUSION In concurrent MetS and CKD, dietary Mg supplementation reduced inflammation and oxidative stress and improved vascular function.
Collapse
|
16
|
Heidari B, Zolfaghari MR, Khademvatani K, Fattahi A, Zarezadeh R. Interrelation among exercise training, cardiac hypertrophy, and tissue kallikrein-kinin system in athlete and non-athlete women. J Cardiovasc Thorac Res 2022; 14:159-165. [DOI: 10.34172/jcvtr.2022.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 08/16/2022] [Indexed: 11/06/2022] Open
Abstract
Introduction: The tissue kallikrein-kinin system is an endogenous homeostatic pathway, which its stimulation is associated with cardioprotection. The present study aimed to determine the effect of exercise training on plasma tissue kallikrein (TK) and bradykinin (BK) and their association with cardiac hypertrophy. Methods: 22 non-athlete and 22 athlete women were exposed to acute (Bruce test) and chronic (12-week swimming training) exercises. 2D echocardiography was used to evaluate morphological and functional features of the heart. Plasma concentrations of TK and BK were quantified by ELISA. Results: Athletes had significantly higher values of left ventricle end-diastolic diameter index (LVEDDI) and left ventricle mass index (LVMI) than non-athletes. Exercise intervention affected echocardiographic features in neither of the study groups. Chronic exercise training notably increased plasma levels of TK and BK, which increase was more pronounced in the athletes. Plasma TK negatively correlated with LVEDDI (r=−0.64, P=0.036 and r=−0.58, P=0.027) and LVMI (r=−0.51, P=0.032 and r=−0.63, P=0.028) in the non-athlete and athlete groups. In opposition, there was a positive correlation between plasma TK and left ventricle ejection fraction in non-athletes (r=0.39, P=0.049) and athletes (r=0.53, P=0.019). Conclusion: The upregulation of the tissue kallikrein-kinin system may be a protective mechanism against excessive cardiac hypertrophy induced by chronic exercise training.
Collapse
Affiliation(s)
- Behnam Heidari
- Department of Physical Education, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| | | | - Kamal Khademvatani
- Cardiology Department, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Abeledo-Machado A, Peña-Zanoni M, Bornancini D, Camilletti MA, Faraoni EY, Marcial A, Rulli S, Alhenc-Gelas F, Díaz-Torga GS. Sex-specific Regulation of Prolactin Secretion by Pituitary Bradykinin Receptors. Endocrinology 2022; 163:6648127. [PMID: 35863039 DOI: 10.1210/endocr/bqac108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Indexed: 11/19/2022]
Abstract
Sex differences in the control of prolactin secretion are well documented. Sex-related differences in intrapituitary factors regulating lactotroph function have recently attracted attention. Sex differences in prolactinoma development are well documented in clinic, prolactinomas being more frequent in women but more aggressive in men, for poorly understood reasons. Kallikrein, the enzyme releasing kinins has been found in the pituitary, but there is no information on pituitary kinin receptors and their function. In the present work, we characterized pituitary bradykinin receptors (BRs) at the messenger RNA and protein levels in 2 mouse models of prolactinoma, Drd2 receptor gene inactivation and hCGβ gene overexpression, in both males and females, wild type or genomically altered. BR B2 (B2R) accounted for 97% or more of total pituitary BRs in both models, regardless of genotype, and was present in lactotrophs, somatotrophs, and gonadotrophs. Male pituitaries displayed higher level of B2R than females, regardless of genotype. Pituitary B2R gene expression was downregulated by estrogen in both males and females but only in females by dopamine. Activation of B1R or B2R by selective pharmacological agonists induced prolactin release in male pituitaries but inhibited prolactin secretion in female pituitaries. Increased B2R content was observed in pituitaries of mutated animals developing prolactinomas, compared to their respective wild-type controls. The present study documents a novel sex-related difference in the control of prolactin secretion and suggests that kinins are involved, through B2R activation, in lactotroph function and prolactinoma development.
Collapse
Affiliation(s)
- Alejandra Abeledo-Machado
- Laboratorio de Fisio-Patología Hormonal, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Milagros Peña-Zanoni
- Laboratorio de Fisio-Patología Hormonal, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Dana Bornancini
- Laboratorio de Fisio-Patología Hormonal, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - María Andrea Camilletti
- Laboratorio de Fisio-Patología Hormonal, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Erika Yanil Faraoni
- Laboratorio de Fisio-Patología Hormonal, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Agustina Marcial
- Laboratorio de Endocrinología Molecular de la Reproducción, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Susana Rulli
- Laboratorio de Endocrinología Molecular de la Reproducción, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Francois Alhenc-Gelas
- INSERM U1138, Universite Paris-Cite, Sorbonne Universite, Centre de Recherche des Cordeliers, Paris, France
| | - Graciela Susana Díaz-Torga
- Laboratorio de Fisio-Patología Hormonal, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| |
Collapse
|
18
|
Edwards A, Kurtcuoglu V. Renal blood flow and oxygenation. Pflugers Arch 2022; 474:759-770. [PMID: 35438336 PMCID: PMC9338895 DOI: 10.1007/s00424-022-02690-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
Our kidneys receive about one-fifth of the cardiac output at rest and have a low oxygen extraction ratio, but may sustain, under some conditions, hypoxic injuries that might lead to chronic kidney disease. This is due to large regional variations in renal blood flow and oxygenation, which are the prerequisite for some and the consequence of other kidney functions. The concurrent operation of these functions is reliant on a multitude of neuro-hormonal signaling cascades and feedback loops that also include the regulation of renal blood flow and tissue oxygenation. Starting with open questions on regulatory processes and disease mechanisms, we review herein the literature on renal blood flow and oxygenation. We assess the current understanding of renal blood flow regulation, reasons for disparities in oxygen delivery and consumption, and the consequences of disbalance between O2 delivery, consumption, and removal. We further consider methods for measuring and computing blood velocity, flow rate, oxygen partial pressure, and related parameters and point out how limitations of these methods constitute important hurdles in this area of research. We conclude that to obtain an integrated understanding of the relation between renal function and renal blood flow and oxygenation, combined experimental and computational modeling studies will be needed.
Collapse
Affiliation(s)
- Aurelie Edwards
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA, 02215, USA
| | - Vartan Kurtcuoglu
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
- National Center of Competence in Research, Kidney.CH, University of Zurich, Zurich, Switzerland.
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
19
|
Nies MK, Yang J, Griffiths M, Damico R, Zhu J, Vaydia D, Fu Z, Brandal S, Austin ED, Ivy DD, Hassoun PM, Van Eyk JE, Everett AD. Proteomics discovery of pulmonary hypertension biomarkers: Insulin-like growth factor binding proteins are associated with disease severity. Pulm Circ 2022; 12:e12039. [PMID: 35514776 PMCID: PMC9063962 DOI: 10.1002/pul2.12039] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/09/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by sustained elevations of pulmonary artery pressure. To date, we lack circulating, diagnostic, and prognostic markers that correlate to clinical and functional parameters. In this study, we performed mass spectrometry-based proteomics analysis to identify circulating biomarkers of PAH. Plasma samples from patients with idiopathic pulmonary arterial hypertension (IPAH, N = 9) and matched normal controls (N = 9) were digested with trypsin and analyzed using data-dependent acquisition on an Orbitrap mass spectrometer. A total of 826 (false discovery rate [FDR] 0.047) and 461 (FDR 0.087) proteins were identified across all plasma samples obtained from IPAH and control subjects, respectively. Of these, 153 proteins showed >2 folds change (p < 0.05) between groups. Circulating levels of carbonic anhydrase 2 (CA2), plasma kallikrein (KLKB1), and the insulin-like growth factor binding proteins (IGFBP1-7) were quantified by immunoassay in an independent verification cohort (N = 36 PAH and N = 35 controls). CA2 and KLKB1 were significantly different in PAH versus control but were not associated with any functional or hemodynamic measurements. Whereas, IGFBP1 and 2 were associated with higher pulmonary vascular resistance, IGFBP2, 4, and 7 with decreased 6-min walk distance (6MWD), and IGFBP1, 2, 4, and 7 with worse survival. This plasma proteomic discovery analysis suggests the IGF axis may serve as important new biomarkers for PAH and play an important role in PAH pathogenesis.
Collapse
Affiliation(s)
- Melanie K. Nies
- Department of Pediatrics, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jun Yang
- Department of Pediatrics, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Megan Griffiths
- Department of Pediatrics, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
- Department of Pediatrics, Division of Pediatric CardiologyColumbia UniversityNew YorkNew YorkUSA
| | - Rachel Damico
- Department of Medicine, Division of Pulmonary and Critical Care MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jie Zhu
- Department of Pediatrics, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Dhananjay Vaydia
- Department of Medicine, Division of Pulmonary and Critical Care MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
- Department of Epidemiology, School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Zongming Fu
- Department of Pediatrics, Division of HematologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Stephanie Brandal
- Department of Pediatrics, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Eric D. Austin
- Department of Pediatrics, Division of Allergy, Immunology, and Pulmonary MedicineVanderbilt UniversityNashvilleTennesseeUSA
| | - Dunbar D. Ivy
- Department of Pediatric CardiologyChildren's Hospital ColoradoAuroraColoradoUSA
| | - Paul M. Hassoun
- Department of Medicine, Division of Pulmonary and Critical Care MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jennifer E. Van Eyk
- Department of Internal Medicine, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
- Advanced Clinical Biosystems Research InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Allen D. Everett
- Department of Pediatrics, Division of CardiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
20
|
Basuli D, Parekh RU, White A, Thayyil A, Sriramula S. Kinin B1 Receptor Mediates Renal Injury and Remodeling in Hypertension. Front Med (Lausanne) 2022; 8:780834. [PMID: 35118089 PMCID: PMC8804098 DOI: 10.3389/fmed.2021.780834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/23/2021] [Indexed: 11/13/2022] Open
Abstract
Despite many readily available therapies, hypertensive kidney disease remains the second most prevalent cause of end-stage renal disease after diabetes, and continues to burden patient populations and escalate morbidity and mortality rates. Kinin B1 receptor (B1R) activation has been shown to have a role in the development of hypertension, one of the major etiologies for chronic kidney disease. However, the role of B1R in hypertension induced renal injury and remodeling remains unexplored. Using a DOCA-salt-induced hypertensive mouse model, we investigated whether B1R deficiency reduces hypertensive renal injury and fibrosis. To further recognize the translational role of B1R, we examined the expression of B1R and its correlation with collagen deposition in renal biopsies from control and hypertensive kidney disease patients. Our data indicates that renal B1R expression was upregulated in the kidneys of DOCA-salt hypertensive mice. Genetic ablation of B1R protected the mice from DOCA-salt-induced renal injury and fibrosis by preventing inflammation and oxidative stress in the kidney. Cultured human proximal tubular epithelial cells expressed B1R and stimulation of B1R with an agonist resulted in increased oxidative stress. In human kidney biopsy samples, we found that the B1R immunoreactivity was not only significantly increased in hypertensive patients compared to normotensive patients, but also there is a positive correlation between B1R expression and renal fibrosis levels. Taken together, our results identify a critical role of B1R in the development of inflammation and fibrosis of the kidney in hypertension.
Collapse
Affiliation(s)
- Debargha Basuli
- Department of Pharmacology and Toxicology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
- Department of Nephrology and Hypertension, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Rohan Umesh Parekh
- Department of Pharmacology and Toxicology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Acacia White
- Department of Pharmacology and Toxicology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Abdullah Thayyil
- Department of Pathology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Srinivas Sriramula
- Department of Pharmacology and Toxicology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
- *Correspondence: Srinivas Sriramula
| |
Collapse
|
21
|
Alves SAS, Florentino LS, Teixeira DE, Silva-Aguiar RP, Peruchetti DB, Oliveira AC, Scharfstein J, Marzolo MP, Pinheiro AAS, Caruso-Neves C. Surface megalin expression is a target to the inhibitory effect of bradykinin on the renal albumin endocytosis. Peptides 2021; 146:170646. [PMID: 34500007 DOI: 10.1016/j.peptides.2021.170646] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/31/2022]
Abstract
Megalin-mediated albumin endocytosis plays a critical role in albumin reabsorption in proximal tubule (PT) epithelial cells (PTECs). Some studies have pointed out the modulatory effect of bradykinin (BK) on urinary protein excretion, but its role in PT protein endocytosis has not yet been determined. Here, we studied the possible correlation between BK and albumin endocytosis in PT. Using LLC-PK1 cells, a model of PTECs, we showed that BK specifically inhibited megalin-mediated albumin endocytosis. This inhibitory effect of BK was mediated by B2 receptor (B2R) because it was abolished by HOE140, an antagonist of B2R, but it was not affected by Lys-des-Arg9-BK, an antagonist of B1. BK induced the stall of megalin in EEA1+ endosomes, but not in LAMP1+ lysosomes, leading to a decrease in surface megalin expression. In addition, we showed that BK, through B2R, activated calphostin C-sensitive protein kinase C, which mediated its effect on the surface megalin expression and albumin endocytosis. These results reveal an important modulatory mechanism of PT albumin endocytosis by BK, which opens new possibilities to understanding the effect of BK on urinary albumin excretion.
Collapse
Affiliation(s)
- Sarah A S Alves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucas S Florentino
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas E Teixeira
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo P Silva-Aguiar
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo B Peruchetti
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Carolina Oliveira
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julio Scharfstein
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - María-Paz Marzolo
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ana Acacia S Pinheiro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.
| |
Collapse
|
22
|
Khodabakhsh P, Asgari Taei A, Mohseni M, Bahrami Zanjanbar D, Khalili H, Masoumi K, Haji Abbas Shirazi A, Dargahi L. Vasoactive Peptides: Role in COVID-19 Pathogenesis and Potential Use as Biomarkers and Therapeutic Targets. Arch Med Res 2021; 52:777-787. [PMID: 34134920 PMCID: PMC8179120 DOI: 10.1016/j.arcmed.2021.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/28/2021] [Accepted: 05/27/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND The ongoing outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), as the latest threat to global health, causes overwhelming effects for the public healthcare systems worldwide. Of note, in addition to the respiratory complications, some patients with coronavirus disease 2019 (COVID-19) also develop serious cardiovascular injuries. Vasoactive peptides play an important role in a wide range of physiological and pathological conditions. AIM With the urgent need for exploring the specific therapeutic targets and biomarkers for the emerging COVID-19, the general aim of this review is to discuss the potentials of the vasoactive peptides including Angiotensin II (Ang II), vasoactive intestinal peptide (VIP), endothelin-1 (ET-1), calcitonin gene-related peptide (CGRP), natriuretic peptides, substance P (SP) and bradykinin (BK) as therapeutic targets and/or prognostic indicators for the COVID-19 pandemic. CONCLUSION Based on various observations some authors conclude that the assessment of vasoactive peptides shall be considered a routine part of COVID-19 patient monitoring, and they can serve as potential therapeutic targets for the disease management.
Collapse
Affiliation(s)
- Pariya Khodabakhsh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran,Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Asgari Taei
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Moein Mohseni
- Pharmaceutical Sciences Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Dorsa Bahrami Zanjanbar
- Pharmaceutical Sciences Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hasti Khalili
- Pharmaceutical Sciences Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kimia Masoumi
- Pharmaceutical Sciences Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Santiago-Hernandez A, Martin-Lorenzo M, Martínez PJ, Gómez-Serrano M, Lopez JA, Cannata P, Esteban V, Heredero A, Aldamiz-Echevarria G, Vázquez J, Ruiz-Hurtado G, Barderas MG, Segura J, Ruilope LM, Alvarez-Llamas G. Early renal and vascular damage within the normoalbuminuria condition. J Hypertens 2021; 39:2220-2231. [PMID: 34261953 DOI: 10.1097/hjh.0000000000002936] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE A continuous association between albuminuria and cardiorenal risk exists further below moderately increased albuminuria ranges. If only based in albumin to creatinine ratio (ACR) higher than 30 mg/g, a significant percentage of individuals may be out of the scope for therapeutic management. Despite epidemiological outcomes, the identification of biochemical changes linked to early albuminuria is underexplored, and normoalbuminuric individuals are usually considered at no risk in clinical practice. Here, we aimed to identify early molecular alterations behind albuminuria development. METHODS Hypertensive patients under renin-angiotensin system (RAS) suppression were classified as control, (ACR < 10 mg/g) or high-normal (ACR = 10-30 mg/g). Urinary protein alterations were quantified and confirmed by untargeted and targeted mass spectrometry. Coordinated protein responses with biological significance in albuminuria development were investigated. Immunohistochemistry assays were performed in human kidney and arterial tissue to in situ evaluate the associated damage. RESULTS A total of 2663 identified proteins reflect inflammation, immune response, ion transport and lipids metabolism (P value ≤ 0.01). A1AT, VTDB and KNG1 varied in high-normal individuals (P value < 0.05), correlated with ACR and associated with the high-normal condition (odds ratio of 20.76, 6.00 and 7.04 were found, respectively (P value < 0.001)). After 12 months, protein variations persist and aggravate in progressors to moderately increased albuminuria. At tissue level, differential protein expression was found in kidney from individuals with moderately increased albuminuria and atherosclerotic aortas for the three proteins, confirming their capacity to reflect subclinical organ damage. CONCLUSION Early renal and vascular damage is molecularly evidenced within the normoalbuminuria condition.
Collapse
Affiliation(s)
| | - Marta Martin-Lorenzo
- Immunoallergy and Proteomics Laboratory, Department of Immunology, IIS-Fundación Jiménez Díaz, UAM
| | - Paula J Martínez
- Immunoallergy and Proteomics Laboratory, Department of Immunology, IIS-Fundación Jiménez Díaz, UAM
| | - María Gómez-Serrano
- Laboratory of Cardiovascular Proteomics, CNIC
- Institute for Tumor Immunology, Philipps University of Marburg, Marburg, Germany
| | - Juan Antonio Lopez
- Laboratory of Cardiovascular Proteomics, CNIC
- CIBER de Enfermedades Cardiovasculares (CIBERCV)
| | | | - Vanesa Esteban
- Department of Allergy and Immunology, IIS-Fundación Jiménez Díaz, UAM
- Red de asma, reacciones adversas y alérgicas (ARADyAL)
- Faculty of Medicine and Biomedicine, Alfonso X El Sabio University
| | | | | | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics, CNIC
- CIBER de Enfermedades Cardiovasculares (CIBERCV)
| | - Gema Ruiz-Hurtado
- CIBER de Enfermedades Cardiovasculares (CIBERCV)
- Cardiorenal Translational Laboratory, Hospital Universitario 12 de Octubre, Madrid
| | - Maria G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos SESCAM, Toledo
| | - Julian Segura
- Hypertension Unit, Hospital Universitario 12 de Octubre
| | - Luis M Ruilope
- Cardiorenal Translational Laboratory, Hospital Universitario 12 de Octubre, Madrid
| | - Gloria Alvarez-Llamas
- Immunoallergy and Proteomics Laboratory, Department of Immunology, IIS-Fundación Jiménez Díaz, UAM
- Red de Investigación Renal (REDINREN), Madrid, Spain
| |
Collapse
|
24
|
Torino C, Tripepi R, Versace MC, Vilasi A, Tripepi G, Panuccio V. Clinical Epidemiology of Systolic and Diastolic Orthostatic Hypotension in Patients on Peritoneal Dialysis. J Clin Med 2021; 10:3075. [PMID: 34300240 PMCID: PMC8304693 DOI: 10.3390/jcm10143075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/29/2021] [Accepted: 07/08/2021] [Indexed: 11/17/2022] Open
Abstract
Blood pressure changes upon standing reflect a hemodynamic response, which depends on the baroreflex system and euvolemia. Dysautonomia and fluctuations in blood volume are hallmarks in kidney failure requiring replacement therapy. Orthostatic hypotension has been associated with mortality in hemodialysis patients, but neither this relationship nor the impact of changes in blood pressure has been tested in patients on peritoneal dialysis. We investigated both these relationships in a cohort of 137 PD patients. The response to orthostasis was assessed according to a standardized protocol. Twenty-five patients (18%) had systolic orthostatic hypotension, and 17 patients (12%) had diastolic hypotension. The magnitude of systolic and diastolic BP changes was inversely related to the value of the corresponding supine BP component (r = -0.16, p = 0.056 (systolic) and r = -0.25, p = 0.003 (diastolic), respectively). Orthostatic changes in diastolic, but not in systolic, BP were linearly related to the death risk (HR (1 mmHg reduction): 1.04, 95% CI 1.01-1.07, p = 0.006), and this was also true for CV death (HR: 1.08, 95% CI 1.03-1.12, p = 0.001). The strength of this association was not affected by further data adjustment (p ≤ 0.05). These findings suggest that independent of the formal diagnosis of orthostatic hypotension, even minor orthostatic reductions in diastolic BP bear an excess death risk in this population.
Collapse
Affiliation(s)
- Claudia Torino
- National Research Council—Institute of Clinical Physiology, Via Vallone Petrara snc, 89124 Reggio Calabria, Italy; (C.T.); (R.T.); (M.C.V.); (A.V.); (G.T.)
| | - Rocco Tripepi
- National Research Council—Institute of Clinical Physiology, Via Vallone Petrara snc, 89124 Reggio Calabria, Italy; (C.T.); (R.T.); (M.C.V.); (A.V.); (G.T.)
| | - Maria Carmela Versace
- National Research Council—Institute of Clinical Physiology, Via Vallone Petrara snc, 89124 Reggio Calabria, Italy; (C.T.); (R.T.); (M.C.V.); (A.V.); (G.T.)
| | - Antonio Vilasi
- National Research Council—Institute of Clinical Physiology, Via Vallone Petrara snc, 89124 Reggio Calabria, Italy; (C.T.); (R.T.); (M.C.V.); (A.V.); (G.T.)
| | - Giovanni Tripepi
- National Research Council—Institute of Clinical Physiology, Via Vallone Petrara snc, 89124 Reggio Calabria, Italy; (C.T.); (R.T.); (M.C.V.); (A.V.); (G.T.)
| | - Vincenzo Panuccio
- Nephology, Dialysis and Transplantation Unit—GOM “Bianchi-Melacrino-Morelli”, Via Vallone Petrara snc, 89124 Reggio Calabria, Italy
| |
Collapse
|
25
|
Beyerstedt S, Casaro EB, Rangel ÉB. COVID-19: angiotensin-converting enzyme 2 (ACE2) expression and tissue susceptibility to SARS-CoV-2 infection. Eur J Clin Microbiol Infect Dis 2021; 40:905-919. [PMID: 33389262 PMCID: PMC7778857 DOI: 10.1007/s10096-020-04138-6] [Citation(s) in RCA: 481] [Impact Index Per Article: 120.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 12/20/2020] [Indexed: 02/06/2023]
Abstract
COVID-19 pandemic is caused by the novel coronavirus SARS-CoV-2. Angiotensin-converting enzyme 2 (ACE2) is not only an enzyme but also a functional receptor on cell surfaces through which SARS-CoV-2 enters the host cells and is highly expressed in the heart, kidneys, and lungs and shed into the plasma. ACE2 is a key regulator of the renin-angiotensin-aldosterone system (RAAS). SARS-CoV-2 causes ACE/ACE2 balance disruption and RAAS activation, which leads ultimately to COVID-19 progression, especially in patients with comorbidities, such as hypertension, diabetes mellitus, and cardiovascular disease. Therefore, ACE2 expression may have paradoxical effects, aiding SARS-CoV-2 pathogenicity, yet conversely limiting viral infection. This article reviews the existing literature and knowledge of ACE2 in COVID-19 setting and focuses on its pathophysiologic involvement in disease progression, clinical outcomes, and therapeutic potential.
Collapse
Affiliation(s)
- Stephany Beyerstedt
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, 627 Albert Einstein Avenue, Building A, Morumbi, São Paulo, SP, Brazil
| | - Expedito Barbosa Casaro
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, 627 Albert Einstein Avenue, Building A, Morumbi, São Paulo, SP, Brazil
| | - Érika Bevilaqua Rangel
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, 627 Albert Einstein Avenue, Building A, Morumbi, São Paulo, SP, Brazil.
- Nephrology Division, Federal University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
26
|
Oz M, Lorke DE. Multifunctional angiotensin converting enzyme 2, the SARS-CoV-2 entry receptor, and critical appraisal of its role in acute lung injury. Biomed Pharmacother 2021; 136:111193. [PMID: 33461019 PMCID: PMC7836742 DOI: 10.1016/j.biopha.2020.111193] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/15/2020] [Accepted: 12/26/2020] [Indexed: 12/11/2022] Open
Abstract
The recent emergence of coronavirus disease-2019 (COVID-19) as a pandemic affecting millions of individuals has raised great concern throughout the world, and the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) was identified as the causative agent for COVID-19. The multifunctional protein angiotensin converting enzyme 2 (ACE2) is accepted as its primary target for entry into host cells. In its enzymatic function, ACE2, like its homologue ACE, regulates the renin-angiotensin system (RAS) critical for cardiovascular and renal homeostasis in mammals. Unlike ACE, however, ACE2 drives an alternative RAS pathway by degrading Ang-II and thus operates to balance RAS homeostasis in the context of hypertension, heart failure, and cardiovascular as well as renal complications of diabetes. Outside the RAS, ACE2 hydrolyzes key peptides, such as amyloid-β, apelin, and [des-Arg9]-bradykinin. In addition to its enzymatic functions, ACE2 is found to regulate intestinal amino acid homeostasis and the gut microbiome. Although the non-enzymatic function of ACE2 as the entry receptor for SARS-CoV-2 has been well established, the contribution of enzymatic functions of ACE2 to the pathogenesis of COVID-19-related lung injury has been a matter of debate. A complete understanding of this central enzyme may begin to explain the various symptoms and pathologies seen in SARS-CoV-2 infected individuals, and may aid in the development of novel treatments for COVID-19.
Collapse
Affiliation(s)
- Murat Oz
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Safat 13110, Kuwait.
| | - Dietrich Ernst Lorke
- Department of Anatomy and Cellular Biology, Khalifa University, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
27
|
Kinins and Kinin Receptors in Cardiovascular and Renal Diseases. Pharmaceuticals (Basel) 2021; 14:ph14030240. [PMID: 33800422 PMCID: PMC8000381 DOI: 10.3390/ph14030240] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/11/2022] Open
Abstract
This review addresses the physiological role of the kallikrein–kinin system in arteries, heart and kidney and the consequences of kallikrein and kinin actions in diseases affecting these organs, especially ischemic and diabetic diseases. Emphasis is put on pharmacological and genetic studies targeting kallikrein; ACE/kininase II; and the two kinin receptors, B1 (B1R) and B2 (B2R), distinguished through the work of Domenico Regoli and his collaborators. Potential therapeutic interest and limitations of the pharmacological manipulation of B1R or B2R activity in cardiovascular and renal diseases are discussed. This discussion addresses either the activation or inhibition of these receptors, based on recent clinical and experimental studies.
Collapse
|
28
|
Dobrek L. An Outline of Renal Artery Stenosis Pathophysiology-A Narrative Review. Life (Basel) 2021; 11:life11030208. [PMID: 33799957 PMCID: PMC8000991 DOI: 10.3390/life11030208] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Renal artery stenosis (RAS) is conditioned mainly by two disturbances: fibromuscular dysplasia or atherosclerosis of the renal artery. RAS is an example of renovascular disease, with complex pathophysiology and consequences. There are multiple pathophysiological mechanisms triggered in response to significant renal artery stenosis, including disturbances within endothelin, kinin-kallikrein and sympathetic nervous systems, with angiotensin II and the renin-angiotensin-aldosterone system (RAAS) playing a central and key role in the pathogenesis of RAS. The increased oxidative stress and the release of pro-inflammatory mediators contributing to pathological tissue remodelling and renal fibrosis are also important pathogenetic elements of RAS. This review briefly summarises these pathophysiological issues, focusing on renovascular hypertension and ischemic nephropathy as major clinical manifestations of RAS. The activation of RAAS and its haemodynamic consequences is the primary and key element in the pathophysiological cascade triggered in response to renal artery stenosis. However, the pathomechanism of RAS is more complex and also includes other disturbances that ultimately contribute to the development of the diseases mentioned above. To sum up, RAS is characterised by different clinical pictures, including asymptomatic disorders diagnosed in kidney imaging, renovascular hypertension, usually characterised by severe course, and chronic ischemic nephropathy, described by pathological remodelling of kidney tissue, ultimately leading to kidney injury and chronic kidney disease.
Collapse
Affiliation(s)
- Lukasz Dobrek
- Department of Clinical Pharmacology, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
29
|
Association of Ang-(1–7) and des-Arg9BK as new biomarkers of obesity and cardiometabolic risk factors in adolescents. Hypertens Res 2021; 44:969-977. [DOI: 10.1038/s41440-021-00618-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/26/2020] [Accepted: 12/23/2020] [Indexed: 11/09/2022]
|
30
|
Wang J, Xia M, Tang X, Jia Z, Li C, Li M, Yin Y, Guo C, Shi J, Liu X, Chen W, Chen T, Feng H. Inhibition of plasma kallikrein mitigates experimental hypertension-enhanced cerebral hematoma expansion. Brain Res Bull 2021; 170:49-57. [PMID: 33556561 DOI: 10.1016/j.brainresbull.2021.02.004] [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: 10/26/2020] [Revised: 01/25/2021] [Accepted: 02/02/2021] [Indexed: 10/22/2022]
Abstract
RATIONALE Hematoma expansion (HE) aggravates brain injury after intracerebral hemorrhage (ICH) and hypertension is a key contributor to HE. Plasma kallikrein (PK) is involved in hemorrhagic transformation in ischemic stroke mice. This study was conducted to explore the role of PK in HE in hypertensive ICH. METHODS Hypertension was achieved by continuous infusion of angiotensin II (Ang II) with an osmotic pump in C57BL/6 mice. ICH was achieved by stereotactic intrastriatal injection of blood. PK-specific antibody and platelet glycoprotein VI (GPVI) agonists were administered to intervene in hematoma expansion. The hematoma volume was indicated by the erythrocyte components hemoglobin and carbonic anhydrase-1 in the ipsilateral brain hemisphere. RESULTS Ang II-induced hypertensive mice showed enhanced hematoma expansion and worsened neurologic deficits after ICH modeling. Moreover, intrastriatal injection of blood from Ang II-treated mice into normal mice increased the area of secondary hemorrhage more than blood from untreated mice. Mechanistically, elevated PK was found in Ang II-infused mice whereas, inhibition of PK and administration of the GPVI agonist convulxin decreased hematoma expansion and improved neurologic deficits after ICH. CONCLUSIONS These findings suggest that PK inhibition and GPVI agonist treatment might serve as potential methods to intervene in HE after ICH.
Collapse
Affiliation(s)
- Jie Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Min Xia
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiaoqin Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhengcai Jia
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chengcheng Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Mingxi Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yi Yin
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chao Guo
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiantao Shi
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xin Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Weixiang Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Tunan Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
31
|
Abassi Z, Skorecki K, Hamo-Giladi DB, Kruzel-Davila E, Heyman SN. Kinins and chymase: the forgotten components of the renin-angiotensin system and their implications in COVID-19 disease. Am J Physiol Lung Cell Mol Physiol 2021; 320:L422-L429. [PMID: 33404363 PMCID: PMC7938643 DOI: 10.1152/ajplung.00548.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The unique clinical features of COVID-19 disease present a formidable challenge in the understanding of its pathogenesis. Within a very short time, our knowledge regarding basic physiological pathways that participate in SARS-CoV-2 invasion and subsequent organ damage have been dramatically expanded. In particular, we now better understand the complexity of the renin-angiotensin-aldosterone system (RAAS) and the important role of angiotensin converting enzyme (ACE)-2 in viral binding. Furthermore, the critical role of its major product, angiotensin (Ang)-(1-7), in maintaining microcirculatory balance and in the control of activated proinflammatory and procoagulant pathways, generated in this disease, have been largely clarified. The kallikrein-bradykinin (BK) system and chymase are intensively interwoven with RAAS through many pathways with complex reciprocal interactions. Yet, so far, very little attention has been paid to a possible role of these physiological pathways in the pathogenesis of COVID-19 disease, even though BK and chymase exert many physiological changes characteristic to this disorder. Herein, we outline the current knowledge regarding the reciprocal interactions of RAAS, BK, and chymase that are probably turned-on in COVID-19 disease and participate in its clinical features. Interventions affecting these systems, such as the inhibition of chymase or blocking BKB1R/BKB2R, might be explored as potential novel therapeutic strategies in this devastating disorder.
Collapse
Affiliation(s)
- Zaid Abassi
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Department of Laboratory Medicine, Rambam Health Care Campus, Haifa, Israel
| | - Karl Skorecki
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Dalit B Hamo-Giladi
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Etty Kruzel-Davila
- Department of Nephrology, Rambam Health Care Campus, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Samuel N Heyman
- Department of Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| |
Collapse
|
32
|
Nonclassical Axis of the Renin-Angiotensin System and Neprilysin: Key Mediators That Underlie the Cardioprotective Effect of PPAR-Alpha Activation during Myocardial Ischemia in a Metabolic Syndrome Model. PPAR Res 2020; 2020:8894525. [PMID: 33354204 PMCID: PMC7737465 DOI: 10.1155/2020/8894525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/21/2022] Open
Abstract
The activation of the renin-angiotensin system (RAS) participates in the development of metabolic syndrome (MetS) and in heart failure. PPAR-alpha activation by fenofibrate reverts some of the effects caused by these pathologies. Recently, nonclassical RAS components have been implicated in the pathogenesis of hypertension and myocardial dysfunction; however, their cardiac functions are still controversial. We evaluated if the nonclassical RAS signaling pathways, directed by angiotensin III and angiotensin-(1-7), are involved in the cardioprotective effect of fenofibrate during ischemia in MetS rats. Control (CT) and MetS rats were divided into the following groups: (a) sham, (b) vehicle-treated myocardial infarction (MI-V), and (c) fenofibrate-treated myocardial infarction (MI-F). Angiotensin III and angiotensin IV levels and insulin increased the aminopeptidase (IRAP) expression and decreased the angiotensin-converting enzyme 2 (ACE2) expression in the hearts from MetS rats. Ischemia activated the angiotensin-converting enzyme (ACE)/angiotensin II/angiotensin receptor 1 (AT1R) and angiotensin III/angiotensin IV/angiotensin receptor 4 (AT4R)-IRAP axes. Fenofibrate treatment prevented the damage due to ischemia in MetS rats by favoring the angiotensin-(1-7)/angiotensin receptor 2 (AT2R) axis and inhibiting the angiotensin III/angiotensin IV/AT4R-IRAP signaling pathway. Additionally, fenofibrate downregulated neprilysin expression and increased bradykinin production. These effects of PPAR-alpha activation were accompanied by a reduction in the size of the myocardial infarct and in the activity of serum creatine kinase. Thus, the regulation of the nonclassical axis of RAS forms part of a novel protective effect of fenofibrate in myocardial ischemia.
Collapse
|
33
|
Wicik Z, Eyileten C, Jakubik D, Simões SN, Martins DC, Pavão R, Siller-Matula JM, Postula M. ACE2 Interaction Networks in COVID-19: A Physiological Framework for Prediction of Outcome in Patients with Cardiovascular Risk Factors. J Clin Med 2020; 9:E3743. [PMID: 33233425 PMCID: PMC7700637 DOI: 10.3390/jcm9113743] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/11/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection (coronavirus disease 2019; COVID-19) is associated with adverse outcomes in patients with cardiovascular disease (CVD). The aim of the study was to characterize the interaction between SARS-CoV-2 and Angiotensin-Converting Enzyme 2 (ACE2) functional networks with a focus on CVD. METHODS Using the network medicine approach and publicly available datasets, we investigated ACE2 tissue expression and described ACE2 interaction networks that could be affected by SARS-CoV-2 infection in the heart, lungs and nervous system. We compared them with changes in ACE-2 networks following SARS-CoV-2 infection by analyzing public data of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). This analysis was performed using the Network by Relative Importance (NERI) algorithm, which integrates protein-protein interaction with co-expression networks. We also performed miRNA-target predictions to identify which miRNAs regulate ACE2-related networks and could play a role in the COVID19 outcome. Finally, we performed enrichment analysis for identifying the main COVID-19 risk groups. RESULTS We found similar ACE2 expression confidence levels in respiratory and cardiovascular systems, supporting that heart tissue is a potential target of SARS-CoV-2. Analysis of ACE2 interaction networks in infected hiPSC-CMs identified multiple hub genes with corrupted signaling which can be responsible for cardiovascular symptoms. The most affected genes were EGFR (Epidermal Growth Factor Receptor), FN1 (Fibronectin 1), TP53, HSP90AA1, and APP (Amyloid Beta Precursor Protein), while the most affected interactions were associated with MAST2 and CALM1 (Calmodulin 1). Enrichment analysis revealed multiple diseases associated with the interaction networks of ACE2, especially cancerous diseases, obesity, hypertensive disease, Alzheimer's disease, non-insulin-dependent diabetes mellitus, and congestive heart failure. Among affected ACE2-network components connected with the SARS-Cov-2 interactome, we identified AGT (Angiotensinogen), CAT (Catalase), DPP4 (Dipeptidyl Peptidase 4), CCL2 (C-C Motif Chemokine Ligand 2), TFRC (Transferrin Receptor) and CAV1 (Caveolin-1), associated with cardiovascular risk factors. We described for the first time miRNAs which were common regulators of ACE2 networks and virus-related proteins in all analyzed datasets. The top miRNAs regulating ACE2 networks were miR-27a-3p, miR-26b-5p, miR-10b-5p, miR-302c-5p, hsa-miR-587, hsa-miR-1305, hsa-miR-200b-3p, hsa-miR-124-3p, and hsa-miR-16-5p. CONCLUSION Our study provides a complete mechanistic framework for investigating the ACE2 network which was validated by expression data. This framework predicted risk groups, including the established ones, thus providing reliable novel information regarding the complexity of signaling pathways affected by SARS-CoV-2. It also identified miRNAs that could be used in personalized diagnosis in COVID-19.
Collapse
Affiliation(s)
- Zofia Wicik
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Santo Andre 09606-045, Brazil; (Z.W.); (D.C.M.J.); (R.P.)
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, 02-091 Warsaw, Poland; (C.E.); (D.J.); (M.P.)
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, 02-091 Warsaw, Poland; (C.E.); (D.J.); (M.P.)
| | - Daniel Jakubik
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, 02-091 Warsaw, Poland; (C.E.); (D.J.); (M.P.)
| | - Sérgio N. Simões
- Federal Institute of Education, Science and Technology of Espírito Santo, Serra, Espírito Santo 29056-264, Brazil;
| | - David C. Martins
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Santo Andre 09606-045, Brazil; (Z.W.); (D.C.M.J.); (R.P.)
| | - Rodrigo Pavão
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Santo Andre 09606-045, Brazil; (Z.W.); (D.C.M.J.); (R.P.)
| | - Jolanta M. Siller-Matula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, 02-091 Warsaw, Poland; (C.E.); (D.J.); (M.P.)
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna,1090 Vienna, Austria
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, 02-091 Warsaw, Poland; (C.E.); (D.J.); (M.P.)
| |
Collapse
|
34
|
Karmouty-Quintana H, Thandavarayan RA, Keller SP, Sahay S, Pandit LM, Akkanti B. Emerging Mechanisms of Pulmonary Vasoconstriction in SARS-CoV-2-Induced Acute Respiratory Distress Syndrome (ARDS) and Potential Therapeutic Targets. Int J Mol Sci 2020; 21:E8081. [PMID: 33138181 PMCID: PMC7662604 DOI: 10.3390/ijms21218081] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
The 1918 influenza killed approximately 50 million people in a few short years, and now, the world is facing another pandemic. In December 2019, a novel coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused an international outbreak of a respiratory illness termed coronavirus disease 2019 (COVID-19) and rapidly spread to cause the worst pandemic since 1918. Recent clinical reports highlight an atypical presentation of acute respiratory distress syndrome (ARDS) in COVID-19 patients characterized by severe hypoxemia, an imbalance of the renin-angiotensin system, an increase in thrombogenic processes, and a cytokine release storm. These processes not only exacerbate lung injury but can also promote pulmonary vascular remodeling and vasoconstriction, which are hallmarks of pulmonary hypertension (PH). PH is a complication of ARDS that has received little attention; thus, we hypothesize that PH in COVID-19-induced ARDS represents an important target for disease amelioration. The mechanisms that can promote PH following SARS-CoV-2 infection are described. In this review article, we outline emerging mechanisms of pulmonary vascular dysfunction and outline potential treatment options that have been clinically tested.
Collapse
Affiliation(s)
- Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Divisions of Pulmonary, Critical Care and Sleep Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | | | - Steven P. Keller
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Sandeep Sahay
- Co-Director, Pulmonary Vascular Diseases Center, The Methodist Hospital, Houston, TX 77030, USA;
| | - Lavannya M. Pandit
- Michael E. DeBakey Veterans Affairs Medical Center, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Bindu Akkanti
- Divisions of Pulmonary, Critical Care and Sleep Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| |
Collapse
|
35
|
Hamid S, Rhaleb IA, Kassem KM, Rhaleb NE. Role of Kinins in Hypertension and Heart Failure. Pharmaceuticals (Basel) 2020; 13:E347. [PMID: 33126450 PMCID: PMC7692223 DOI: 10.3390/ph13110347] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
The kallikrein-kinin system (KKS) is proposed to act as a counter regulatory system against the vasopressor hormonal systems such as the renin-angiotensin system (RAS), aldosterone, and catecholamines. Evidence exists that supports the idea that the KKS is not only critical to blood pressure but may also oppose target organ damage. Kinins are generated from kininogens by tissue and plasma kallikreins. The putative role of kinins in the pathogenesis of hypertension is discussed based on human mutation cases on the KKS or rats with spontaneous mutation in the kininogen gene sequence and mouse models in which the gene expressing only one of the components of the KKS has been deleted or over-expressed. Some of the effects of kinins are mediated via activation of the B2 and/or B1 receptor and downstream signaling such as eicosanoids, nitric oxide (NO), endothelium-derived hyperpolarizing factor (EDHF) and/or tissue plasminogen activator (T-PA). The role of kinins in blood pressure regulation at normal or under hypertension conditions remains debatable due to contradictory reports from various laboratories. Nevertheless, published reports are consistent on the protective and mediating roles of kinins against ischemia and cardiac preconditioning; reports also demonstrate the roles of kinins in the cardiovascular protective effects of the angiotensin-converting enzyme (ACE) and angiotensin type 1 receptor blockers (ARBs).
Collapse
Affiliation(s)
- Suhail Hamid
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA; (S.H.); (I.A.R.)
| | - Imane A. Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA; (S.H.); (I.A.R.)
| | - Kamal M. Kassem
- Division of Cardiology, Department of Internal Medicine, University of Louisville Medical Center, Louisville, KY 40202, USA;
| | - Nour-Eddine Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA; (S.H.); (I.A.R.)
- Department of Physiology, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
36
|
Hao P, Liu Y, Guo H, Zhang Z, Chen Q, Hao G, Zhang C, Zhang Y. Prolylcarboxypeptidase Mitigates Myocardial Ischemia/Reperfusion Injury by Stabilizing Mitophagy. Front Cell Dev Biol 2020; 8:584933. [PMID: 33195231 PMCID: PMC7642202 DOI: 10.3389/fcell.2020.584933] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022] Open
Abstract
The role of prolylcarboxypeptidase (PRCP) in myocardial ischemia/reperfusion (I/R) injury is unclear. Herein, we aimed to evaluate the protective effect of the PRCP-angiotensin-(1-7) [Ang-(1-7)]/bradykinin-(1-9) [BK-(1-9)] axis on myocardial I/R injury and identify the mechanisms involved. Plasma PRCP level and activity, as well as Ang-(1-7) and BK-(1-9) levels, were compared in healthy subjects, patients with unstable angina, and those with ST-segment-elevated acute myocardial infarction (AMI). Thereafter, the effects of PRCP overexpression and knockdown on left ventricular function, mitophagy, and levels of Ang-(1-7) and BK-(1-9) were examined in rats during myocardial I/R. Finally, the effects of Ang-(1-7) and BK-(1-9) on I/R-induced mitophagy and the signaling pathways involved were investigated in vitro in rat cardiomyocytes. AMI patients showed increased plasma level and activity of PRCP and levels of Ang-(1-7) and BK-(1-9) as compared with healthy subjects and those with unstable angina. PRCP protected against myocardial I/R injury in rats by paradoxical regulation of cardiomyocyte mitophagy during the ischemia and reperfusion phases, which was mediated by downstream Ang-(1-7) and BK-(1-9). We further depicted a possible role of activation of AMPK in mitophagy induction during ischemia and activation of Akt in mitophagy inhibition during reperfusion in the beneficial effects of Ang-(1-7) and BK-(1-9). Thus, the PRCP-Ang-(1-7)/BK-(1-9) axis may protect against myocardial I/R injury by paradoxical regulation of cardiomyocyte mitophagy during ischemia and reperfusion phases.
Collapse
Affiliation(s)
- Panpan Hao
- Department of Cardiology, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yanping Liu
- Department of Cardiology, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shenzhen Research Institute of Shandong University, Shenzhen, China
| | - Haipeng Guo
- Department of Cardiology, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhongwen Zhang
- Department of Endocrinology and Metabolism, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Shandong University, Jinan, China
| | - Qingjie Chen
- First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Guoxiang Hao
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Cheng Zhang
- Department of Cardiology, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yun Zhang
- Department of Cardiology, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
37
|
Gambardella J, Sorriento D, Bova M, Rusciano M, Loffredo S, Wang X, Petraroli A, Carucci L, Mormile I, Oliveti M, Bruno Morelli M, Fiordelisi A, Spadaro G, Campiglia P, Sala M, Trimarco B, Iaccarino G, Santulli G, Ciccarelli M. Role of Endothelial G Protein-Coupled Receptor Kinase 2 in Angioedema. Hypertension 2020; 76:1625-1636. [PMID: 32895019 DOI: 10.1161/hypertensionaha.120.15130] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Excessive BK (bradykinin) stimulation is responsible for the exaggerated permeabilization of the endothelium in angioedema. However, the molecular mechanisms underlying these responses have not been investigated. BK receptors are Gq-protein-coupled receptors phosphorylated by GRK2 (G protein-coupled receptor kinase 2) with a hitherto unknown biological and pathophysiological significance. In the present study, we sought to identify the functional role of GRK2 in angioedema through the regulation of BK signaling. We found that the accumulation of cytosolic Ca2+ in endothelial cells induced by BK was sensitive to GRK2 activity, as it was significantly augmented by inhibiting the kinase. Accordingly, permeabilization and NO production induced by BK were enhanced, as well. In vivo, mice with reduced GRK2 levels in the endothelium (Tie2-CRE/GRK2fl+/fl-) exhibited an increased response to BK in terms of vascular permeability and extravasation. Finally, patients with reduced GRK2 levels displayed a severe phenotype of angioedema. Taken together, these findings establish GRK2 as a novel pivotal regulator of BK signaling with an essential role in the pathophysiology of vascular permeability and angioedema.
Collapse
Affiliation(s)
- Jessica Gambardella
- From the Department of Advanced Biomedical Science (J.G., D.S., A.F., B.T., G.I., G. Santulli), University of Naples Federico II, NA, Italy.,Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., X.W., M.B.M., G. Santulli), Albert Einstein College of Medicine, Montefiore University Hospital, NY.,Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM) (J.G., X.W., M.B.M., G. Santulli), Albert Einstein College of Medicine, Montefiore University Hospital, NY.,International Translational Research and Medical Education Consortium (ITME), NA, Italy (J.G., B.T., G. Santulli)
| | - Daniela Sorriento
- Department of Translational Medical Sciences and Interdepartmental Center for Research in Basic and Clinical Immunology Sciences (M.B., S.L., A.P., L.C., I.M., G. Spadaro), University of Naples Federico II, NA, Italy
| | - Maria Bova
- Department of Translational Medical Sciences and Interdepartmental Center for Research in Basic and Clinical Immunology Sciences (M.B., S.L., A.P., L.C., I.M., G. Spadaro), University of Naples Federico II, NA, Italy
| | - Mariarosaria Rusciano
- Montevergine Hospital, Mercogliano, Italy (M.R.).,Department of Medicine and Surgery (M.R., M.O., M.C.), University of Salerno, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences and Interdepartmental Center for Research in Basic and Clinical Immunology Sciences (M.B., S.L., A.P., L.C., I.M., G. Spadaro), University of Naples Federico II, NA, Italy
| | - Xujun Wang
- Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., X.W., M.B.M., G. Santulli), Albert Einstein College of Medicine, Montefiore University Hospital, NY.,Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM) (J.G., X.W., M.B.M., G. Santulli), Albert Einstein College of Medicine, Montefiore University Hospital, NY
| | - Angelica Petraroli
- Department of Translational Medical Sciences and Interdepartmental Center for Research in Basic and Clinical Immunology Sciences (M.B., S.L., A.P., L.C., I.M., G. Spadaro), University of Naples Federico II, NA, Italy
| | - Laura Carucci
- Department of Translational Medical Sciences and Interdepartmental Center for Research in Basic and Clinical Immunology Sciences (M.B., S.L., A.P., L.C., I.M., G. Spadaro), University of Naples Federico II, NA, Italy
| | - Ilaria Mormile
- Department of Translational Medical Sciences and Interdepartmental Center for Research in Basic and Clinical Immunology Sciences (M.B., S.L., A.P., L.C., I.M., G. Spadaro), University of Naples Federico II, NA, Italy
| | - Marco Oliveti
- Department of Medicine and Surgery (M.R., M.O., M.C.), University of Salerno, Italy
| | - Marco Bruno Morelli
- Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., X.W., M.B.M., G. Santulli), Albert Einstein College of Medicine, Montefiore University Hospital, NY.,Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM) (J.G., X.W., M.B.M., G. Santulli), Albert Einstein College of Medicine, Montefiore University Hospital, NY
| | - Antonella Fiordelisi
- From the Department of Advanced Biomedical Science (J.G., D.S., A.F., B.T., G.I., G. Santulli), University of Naples Federico II, NA, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences and Interdepartmental Center for Research in Basic and Clinical Immunology Sciences (M.B., S.L., A.P., L.C., I.M., G. Spadaro), University of Naples Federico II, NA, Italy
| | - Pietro Campiglia
- Division of Biomedicine, Department of Pharmaceutical Science (P.C., M.S.), University of Salerno, Italy
| | - Marina Sala
- Division of Biomedicine, Department of Pharmaceutical Science (P.C., M.S.), University of Salerno, Italy
| | - Bruno Trimarco
- International Translational Research and Medical Education Consortium (ITME), NA, Italy (J.G., B.T., G. Santulli)
| | - Guido Iaccarino
- From the Department of Advanced Biomedical Science (J.G., D.S., A.F., B.T., G.I., G. Santulli), University of Naples Federico II, NA, Italy
| | - Gaetano Santulli
- From the Department of Advanced Biomedical Science (J.G., D.S., A.F., B.T., G.I., G. Santulli), University of Naples Federico II, NA, Italy.,Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., X.W., M.B.M., G. Santulli), Albert Einstein College of Medicine, Montefiore University Hospital, NY.,Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM) (J.G., X.W., M.B.M., G. Santulli), Albert Einstein College of Medicine, Montefiore University Hospital, NY.,International Translational Research and Medical Education Consortium (ITME), NA, Italy (J.G., B.T., G. Santulli)
| | - Michele Ciccarelli
- Department of Medicine and Surgery (M.R., M.O., M.C.), University of Salerno, Italy
| |
Collapse
|
38
|
Curran CS, Rivera DR, Kopp JB. COVID-19 Usurps Host Regulatory Networks. Front Pharmacol 2020; 11:1278. [PMID: 32922297 PMCID: PMC7456869 DOI: 10.3389/fphar.2020.01278] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/03/2020] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes coronavirus disease 2019 (COVID-19). SARS-CoV-2 binds the angiotensin-converting enzyme 2 (ACE2) on the cell surface and this complex is internalized. ACE2 serves as an endogenous inhibitor of inflammatory signals associated with four major regulator systems: the renin-angiotensin-aldosterone system (RAAS), the complement system, the coagulation cascade, and the kallikrein-kinin system (KKS). Understanding the pathophysiological effects of SARS-CoV-2 on these pathways is needed, particularly given the current lack of proven, effective treatments. The vasoconstrictive, prothrombotic and pro-inflammatory conditions induced by SARS-CoV-2 can be ascribed, at least in part, to the activation of these intersecting physiological networks. Moreover, patients with immune deficiencies, hypertension, diabetes, coronary heart disease, and kidney disease often have altered activation of these pathways, either due to underlying disease or to medications, and may be more susceptible to SARS-CoV-2 infection. Certain characteristic COVID-associated skin, sensory, and central nervous system manifestations may also be linked to viral activation of the RAAS, complement, coagulation, and KKS pathways. Pharmacological interventions that target molecules along these pathways may be useful in mitigating symptoms and preventing organ or tissue damage. While effective anti-viral therapies are critically needed, further study of these pathways may identify effective adjunctive treatments and patients most likely to benefit.
Collapse
Affiliation(s)
- Colleen S Curran
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Donna R Rivera
- Surveillance Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
39
|
Polidoro RB, Hagan RS, de Santis Santiago R, Schmidt NW. Overview: Systemic Inflammatory Response Derived From Lung Injury Caused by SARS-CoV-2 Infection Explains Severe Outcomes in COVID-19. Front Immunol 2020; 11:1626. [PMID: 32714336 PMCID: PMC7344249 DOI: 10.3389/fimmu.2020.01626] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/17/2020] [Indexed: 01/12/2023] Open
Abstract
Most SARS-CoV2 infections will not develop into severe COVID-19. However, in some patients, lung infection leads to the activation of alveolar macrophages and lung epithelial cells that will release proinflammatory cytokines. IL-6, TNF, and IL-1β increase expression of cell adhesion molecules (CAMs) and VEGF, thereby increasing permeability of the lung endothelium and reducing barrier protection, allowing viral dissemination and infiltration of neutrophils and inflammatory monocytes. In the blood, these cytokines will stimulate the bone marrow to produce and release immature granulocytes, that return to the lung and further increase inflammation, leading to acute respiratory distress syndrome (ARDS). This lung-systemic loop leads to cytokine storm syndrome (CSS). Concurrently, the acute phase response increases the production of platelets, fibrinogen and other pro-thrombotic factors. Systemic decrease in ACE2 function impacts the Renin-Angiotensin-Kallikrein-Kinin systems (RAS-KKS) increasing clotting. The combination of acute lung injury with RAS-KKS unbalance is herein called COVID-19 Associated Lung Injury (CALI). This conservative two-hit model of systemic inflammation due to the lung injury allows new intervention windows and is more consistent with the current knowledge.
Collapse
Affiliation(s)
- Rafael B. Polidoro
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Robert S. Hagan
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | | | - Nathan W. Schmidt
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
40
|
Vio CP, Gallardo P, Cespedes C, Salas D, Diaz-Elizondo J, Mendez N. Dietary Potassium Downregulates Angiotensin-I Converting Enzyme, Renin, and Angiotensin Converting Enzyme 2. Front Pharmacol 2020; 11:920. [PMID: 32625100 PMCID: PMC7314933 DOI: 10.3389/fphar.2020.00920] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 06/05/2020] [Indexed: 12/16/2022] Open
Abstract
Background The importance of dietary potassium in health and disease has been underestimated compared with that placed on dietary sodium. Larger effort has been made on reduction of sodium intake and less on the adequate dietary potassium intake, although natural food contains much more potassium than sodium. The benefits of a potassium-rich diet are known, however, the mechanism by which it exerts its preventive action, remains to be elucidated. With the hypothesis that dietary potassium reduces renal vasoconstrictor components of the renin-angiotensin system in the long-term, we studied the effect of high potassium diet on angiotensin-I converting enzyme, renin, and angiotensin converting enzyme 2. Methods Sprague Dawley male rats on a normal sodium diet received normal potassium (0.9%, NK) or high potassium diet (3%, HK) for 4 weeks. Urine was collected in metabolic cages for electrolytes and urinary volume measurement. Renal tissue was used to analyze angiotensin-I converting enzyme, renin, and angiotensin converting enzyme 2 expression. Protein abundance analysis was done by Western blot; gene expression by mRNA levels by RT-qPCR. Renal distribution of angiotensin-I converting enzyme and renin was done by immunohistochemistry and morphometric analysis in coded samples. Results High potassium diet (4 weeks) reduced the levels of renin, angiotensin-I converting enzyme, and angiotensin converting enzyme 2. Angiotensin-I converting enzyme was located in the brush border of proximal tubules and with HK diet decreased the immunostaining intensity (P < 0.05), decreased the mRNA (P < 0.01) and the protein levels (P < 0.01). Renin localization was restricted to granular cells of the afferent arteriole and HK diet decreased the number of renin positive cells (P < 0.01) and renin mRNA levels (P < 0.01). High potassium intake decreased angiotensin converting enzyme 2 gene expression and protein levels (P < 0.01).No morphological abnormalities were observed in renal tissue during high potassium diet.The reduced expression of angiotensin-I converting enzyme, renin, and angiotensin converting enzyme 2 during potassium supplementation suggest that high dietary potassium intake could modulate these vasoactive enzymes and this effects can contribute to the preventive and antihypertensive effect of potassium.
Collapse
Affiliation(s)
- Carlos P Vio
- Center for Aging and Regeneration CARE UC, Department of Physiology, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastian, Santiago, Chile
| | - Pedro Gallardo
- Facultad de Medicina, Escuela de Medicina, Universidad Finis Terrae, Santiago, Chile
| | - Carlos Cespedes
- Center for Aging and Regeneration CARE UC, Department of Physiology, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastian, Santiago, Chile
| | - Daniela Salas
- Center for Aging and Regeneration CARE UC, Department of Physiology, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jessica Diaz-Elizondo
- Center for Aging and Regeneration CARE UC, Department of Physiology, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Natalia Mendez
- Facultad de Medicina, Institute of Anatomy, Histology and Pathology, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
41
|
Gu L, Wang J, Zhang DD, Meng X, Zhang Y, Zhang J, Zhang H, Guo X, Lin DH, Wang WH, Gu RM. Inhibition of AT2R and Bradykinin Type II Receptor (BK2R) Compromises High K + Intake-Induced Renal K + Excretion. Hypertension 2019; 75:439-448. [PMID: 31865783 DOI: 10.1161/hypertensionaha.119.13852] [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: 11/16/2022]
Abstract
The inhibition of Type II angiotensin II receptor (AT2R) or BK2R (bradykinin type II receptor) stimulates basolateral Kir4.1/Kir5.1 in the distal convoluted tubule (DCT) and activates thiazide-sensitive NCC (Na-Cl cotransporter). The aim of the present study is to examine the role of AT2R and BK2R in mediating the effect of HK (high dietary K+) intake on the basolateral K+ channels, NCC, and renal K+ excretion. Feeding mice (male and female) with HK diet for overnight significantly decreased the basolateral K+ conductance, depolarized the DCT membrane, diminished the expression of pNCC (phosphorylated NCC) and tNCC (total NCC), and decreased thiazide-sensitive natriuresis. Overnight HK intake also increased the expression of cleaved ENaC-α and -γ subunits but had no effect on NKCC2 expression. Pretreatment of the mice (male and female) with PD123319 and HOE140 stimulated the expression of tNCC and pNCC, augmented hydrochlorothiazide-induced natriuresis, and increased the negativity of the DCT membrane. The deletion of Kir4.1 not only decreased the NCC activity but also abolished the stimulatory effect of PD123319 and HOE140 perfusion on NCC activity. Moreover, the effect of overnight HK loading on Kir4.1/Kir5.1 in the DCT and NCC expression/activity was compromised in the mice treated with AT2R/BK2R antagonists. Renal clearance study showed that inhibition of AT2R and BK2R impairs renal K+ excretion in response to overnight HK loading, and the mice pretreated with PD123319 and HOE140 were hyperkalemic during HK intake. We conclude that synergistic activation of AT2R and BK2R is required for the effect of overnight HK diet on Kir4.1/Kir5.1 in the DCT and NCC activity.
Collapse
Affiliation(s)
- Li Gu
- From the Department of Physiology, Harbin Medical University, China (L.G., J.W., X.M., Y.Z, J.Z., H.Z., X.G., R.-M.G.)
| | - JunLin Wang
- From the Department of Physiology, Harbin Medical University, China (L.G., J.W., X.M., Y.Z, J.Z., H.Z., X.G., R.-M.G.)
| | - Dan-Dan Zhang
- Department of Pharmacology, New York Medical College, Valhalla (D.-D. Z., D.-H.L, W.-H.W.)
| | - XinXin Meng
- From the Department of Physiology, Harbin Medical University, China (L.G., J.W., X.M., Y.Z, J.Z., H.Z., X.G., R.-M.G.)
| | - YunHong Zhang
- From the Department of Physiology, Harbin Medical University, China (L.G., J.W., X.M., Y.Z, J.Z., H.Z., X.G., R.-M.G.)
| | - JiaWen Zhang
- From the Department of Physiology, Harbin Medical University, China (L.G., J.W., X.M., Y.Z, J.Z., H.Z., X.G., R.-M.G.)
| | - Hao Zhang
- From the Department of Physiology, Harbin Medical University, China (L.G., J.W., X.M., Y.Z, J.Z., H.Z., X.G., R.-M.G.)
| | - XiWen Guo
- From the Department of Physiology, Harbin Medical University, China (L.G., J.W., X.M., Y.Z, J.Z., H.Z., X.G., R.-M.G.)
| | - Dao-Hong Lin
- Department of Pharmacology, New York Medical College, Valhalla (D.-D. Z., D.-H.L, W.-H.W.)
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College, Valhalla (D.-D. Z., D.-H.L, W.-H.W.)
| | - Rui-Min Gu
- From the Department of Physiology, Harbin Medical University, China (L.G., J.W., X.M., Y.Z, J.Z., H.Z., X.G., R.-M.G.)
| |
Collapse
|
42
|
Associations of ACE I/D polymorphism with the levels of ACE, kallikrein, angiotensin II and interleukin-6 in STEMI patients. Sci Rep 2019; 9:19719. [PMID: 31873176 PMCID: PMC6927979 DOI: 10.1038/s41598-019-56263-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022] Open
Abstract
This study aimed to compare the plasma levels of angiotensin-I converting enzyme (ACE), Angiotensin II (AngII), kallikrein (KLK1) and interleukin-6 (IL-6) in ST segment elevation myocardial infarction (STEMI) patients with different ACE Insertion/deletion (I/D) polymorphisms in a Chinese population. The ACE genotypes were determined in the 199 STEMI patients and 216 control subjects. STEMI patients were divided into three groups based on the ACE genotypes. Single polymerase chain reaction (PCR) was performed to characterize ACE I/D polymorphisms. Plasma levels of ACE, AngII, KLK1 and IL-6 were measured by enzyme-linked immunosorbent assay (ELISA). We found that the DD or ID genotype was significantly independently associated with high ACE (OR = 4.697; 95% CI = 1.927–11.339), KLK1 (3.339; 1.383–8.063) and IL-6 levels (OR = 2.10; 1.025–4.327) in STEMI patients. However, there was no statistical significance between the ACE I/D polymorphism and AngII plasma levels whether in univariate or multivariate logistic regression. Additionally, we detected a significantly positive correlation between plasma KLK1 levels and IL-6 levels in STEMI patients (r = 0.584, P < 0.001). The study showed high levels of ACE, KLK1 and IL-6 were detected when the D allele was present, but AngII plasma levels was not influenced by the ACE I/D polymorphism.
Collapse
|
43
|
Role of protease-activated receptor 2 in regulation of renin synthesis and secretion in mice. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1401-1410. [DOI: 10.1007/s00210-019-01677-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/13/2019] [Indexed: 11/25/2022]
|
44
|
Zhang D, Pollock DM. Diurnal Regulation of Renal Electrolyte Excretion: The Role of Paracrine Factors. Annu Rev Physiol 2019; 82:343-363. [PMID: 31635525 DOI: 10.1146/annurev-physiol-021119-034446] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many physiological processes, including most kidney-related functions, follow specific rhythms tied to a 24-h cycle. This is largely because circadian genes operate in virtually every cell type in the body. In addition, many noncanonical genes have intrinsic circadian rhythms, especially within the liver and kidney. This new level of complexity applies to the control of renal electrolyte excretion. Furthermore, there is growing evidence that paracrine and autocrine factors, especially the endothelin system, are regulated by clock genes. We have known for decades that excretion of electrolytes is dependent on time of day, which could play an important role in fluid volume balance and blood pressure control. Here, we review what is known about the interplay between paracrine and circadian control of electrolyte excretion. The hope is that recognition of paracrine and circadian factors can be considered more deeply in the future when integrating with well-established neuroendocrine control of excretion.
Collapse
Affiliation(s)
- Dingguo Zhang
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35233, USA; ,
| | - David M Pollock
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35233, USA; ,
| |
Collapse
|
45
|
Okamoto R, Ali Y, Hashizume R, Suzuki N, Ito M. BNP as a Major Player in the Heart-Kidney Connection. Int J Mol Sci 2019; 20:ijms20143581. [PMID: 31336656 PMCID: PMC6678680 DOI: 10.3390/ijms20143581] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Brain natriuretic peptide (BNP) is an important biomarker for patients with heart failure, hypertension and cardiac hypertrophy. Although it is known that BNP levels are relatively higher in patients with chronic kidney disease and no heart disease, the mechanism remains unknown. Here, we review the functions and the roles of BNP in the heart-kidney interaction. In addition, we discuss the relevant molecular mechanisms that suggest BNP is protective against chronic kidney diseases and heart failure, especially in terms of the counterparts of the renin-angiotensin-aldosterone system (RAAS). The renal medulla has been reported to express depressor substances. The extract of the papillary tips from kidneys may induce the expression and secretion of BNP from cardiomyocytes. A better understanding of these processes will help accelerate pharmacological treatments for heart-kidney disease.
Collapse
Affiliation(s)
- Ryuji Okamoto
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan.
| | - Yusuf Ali
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Ryotaro Hashizume
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Noboru Suzuki
- Department of Animal Genomics, Functional Genomics Institute, Mie University Life Science Research Center, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Masaaki Ito
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| |
Collapse
|
46
|
Yang M, Zhou N, Zhang H, Kang G, Cao B, Kang Q, Li R, Zhu X, Rao W, Yu Q. Kininogen-1 as a protein biomarker for schizophrenia through mass spectrometry and genetic association analyses. PeerJ 2019; 7:e7327. [PMID: 31346501 PMCID: PMC6642793 DOI: 10.7717/peerj.7327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/19/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Schizophrenia (SCZ) is a complex and severe mental illness. There is a lack of effective biomarkers for SCZ diagnosis. The aim of this study was to explore the possibility of using serum peptides for the diagnosis of SCZ as well as analyze the association of variants in genes coding for these peptides and SCZ. METHODS After bead-based fractionation, the matrix-assisted laser desorption ionization/time-of-flight mass spectrometry technique was used to identify peptides that showed different expressions between 166 SCZ patients and 201 healthy controls. Differentially expressed peptides were verified in a second set of samples (81 SCZ patients and 103 healthy controls). The association of SCZ and three tagSNPs selected in genes coding for differentially expressed peptides was performed in 1,126 SCZ patients and 1,168 controls. RESULTS The expression level of peptides with m/z 1,945.07 was significant lower in SCZ patients than in healthy controls (P < 0.000001). The peptide with m/z 1,945.07 was confirmed to be a fragment of Kininogen-1. In the verification tests, Kininogen-1 had a sensitivity of 95.1% and a specificity of 97.1% in SCZ prediction. Among the three tagSNPs (rs13037490, rs2983639, rs2983640) selected in the Cystatin 9 gene (CST9) which encodes peptides including Kininogen-1, tagSNP rs2983640 had its genotype distributions significantly different between SCZ patients and controls under different genetic models (P < 0.05). Haplotypes CG (rs2983639-rs2983640) and TCG (rs13037490-rs2983639-rs2983640) were significantly associated with SCZ (CG: OR = 1.21, 95% CI [1.02-1.44], P = 0.032; TCG: OR = 24.85, 95% CI [5.98-103.17], P < 0.0001). CONCLUSIONS The present study demonstrated that SCZ patients had decreased expression of Kininogen-1 and genetic variants in Kininogen-1 coding gene CST9 were significantly associated with SCZ. The findings from both protein and genetic association studies suggest that Kininogen-1 could be a biomarker of SCZ.
Collapse
Affiliation(s)
- Mingjia Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Na Zhou
- Department of Pharmacy, Hospital of Stomatology, Jilin University, Changchun, Jilin Province, China
| | - Huiping Zhang
- Department of Psychiatry and Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
| | - Guojun Kang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Bonan Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Qi Kang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Rixin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Xiaojing Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Wenwang Rao
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
- Unit of Psychiatry, Faculty of Health Sciences, University of Macau, Macao, China
| | - Qiong Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
47
|
Combined Antihypertensive Therapies That Increase Expression of Cardioprotective Biomarkers Associated With the Renin-Angiotensin and Kallikrein-Kinin Systems. J Cardiovasc Pharmacol 2019; 72:291-295. [PMID: 30422889 DOI: 10.1097/fjc.0000000000000629] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Antihypertensive pharmacological treatments focus on the use of angiotensin-converting enzyme (ACE) inhibitors, AT1 receptor antagonists, and beta-blockers as single and combined treatments. The effect of single treatments on the mRNA expression of some components of the renin-angiotensin system has been studied, but not the effect of combined treatments. This study determined the expression of the AT1, AT2, B1, and B2 receptors and of the enzymes ACE and ACE2 in hypertensive rats treated with captopril-propranolol or losartan-propranolol. Methods: The mRNA expression of the receptors and enzymes was determined by reverse transcription-quantitative polymerase chain reaction in the aorta of spontaneously hypertensive rats under different treatments. Results: Rats under combined treatments showed a decrease in the expression of AT1 and ACE, and an increase in the expression of the B1 receptor (captopril + propranolol group: 0.43 ± 0.046, 2.243 ± 0.269, 3.356 ± 0.418; Group: losartan + propranolol: 0.727 ± 0.071, 0.852 ± 0.102, 1.277 ± 0.131 compared to the spontaneously hypertensive group: 1 ± 0.212, 1 ± 0.192, 1 ± 0.214). This decrease in the expression of ACE and AT1 suggests a reduction in the expression of Ang II that could be related to a lower response to this vasoconstrictor. An increase in the expression of B1 would improve vasodilation, which would be a beneficial effect of combined therapies for hypertension.
Collapse
|
48
|
Kim H, Lee JK. The effects of combined exercise program on health and physical fitness factors according to a BglI RFLP in renin gene in middle-aged obese women. GAZZETTA MEDICA ITALIANA ARCHIVIO PER LE SCIENZE MEDICHE 2019. [DOI: 10.23736/s0393-3660.18.03853-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
49
|
Insertions of antihypertensive peptides and their applications in pharmacy and functional foods. Appl Microbiol Biotechnol 2019; 103:2493-2505. [DOI: 10.1007/s00253-019-09633-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/06/2019] [Accepted: 01/07/2019] [Indexed: 12/15/2022]
|
50
|
Cao L, Cobbs A, Simon RP, Zhou A. Distinct plasma proteomic changes in male and female African American stroke patients. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2019; 11:12-20. [PMID: 31149323 PMCID: PMC6526384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/05/2019] [Indexed: 03/19/2023]
Abstract
BACKGROUND Stroke occurs more often and results in more severe brain injury in African Americans than in Caucasians. The former also exhibit different responses to thrombolytic therapy than the latter do. There is an imminent need for stroke biomarkers for African Americans, who have been underrepresented in biomarker research for stroke diagnosis and prognosis. Proteomics offers sources for protein biomarkers that are not available by other Omics approaches. In this pilot study, plasma proteomes of African American stroke patients were analyzed and compared to that of hypertensive, non-stroke controls. METHODS Plasma samples were prepared from whole blood specimens that were collected from stroke patients admitted to Grady Memorial Hospital in Atlanta, and their age- and sex-matched, hypertensive controls from the outpatient clinic. Samples were pooled according to patient groups and sex. Plasma proteins were analyzed with quantitative mass spectrometry. The identified and quantified proteins were compared between stroke and control patients of each sex. Proteins that showed changes in abundances in stroke patients were further analyzed with the assistance of bioinformatics tools for their known biological functions or potential implications in stroke. RESULTS A total of 128 annotated proteins were identified. Results of bioinformatic analysis of plasma proteins whose levels were increased in stroke patients showed, as expected, their association with blood coagulation and inflammation processes. Interestingly, a number of proteins showed different or even opposing changes in male and female stroke patients, notably those involved in IL-4 and IL-6 signaling, complement activation, and blood coagulation disorders. For a few proteins that were increased in female but unchanged or decreased in male stroke patients, an association with fibromuscular dysplasia was recognized. CONCLUSION Plasma proteins that differ in quantities between stroke patients and controls were readily detected using a simple proteomic approach. Sex-dependent changes and changes that have not been reported for African American stroke patients offer potentially novel biomarkers for stroke in this underserved population.
Collapse
Affiliation(s)
- Li Cao
- Neuroscience Institute, Morehouse School of Medicine Atlanta, Georgia.,GSK Institute Philadelphia, Pennsylvania
| | - Alyssa Cobbs
- Neuroscience Institute, Morehouse School of Medicine Atlanta, Georgia
| | - Roger P Simon
- Neuroscience Institute, Morehouse School of Medicine Atlanta, Georgia.,Grady Memorial Hospital Atlanta, Georgia
| | - An Zhou
- Neuroscience Institute, Morehouse School of Medicine Atlanta, Georgia
| |
Collapse
|