1
|
Xu S, Wen S, Zong X, Wen S, Zhu J, Zheng W, Wang Z, Cao P, Liang Z, Ding C, Zhang Y, Ruan G. Identification of Circulating Proteins Associated With Blood Pressure. Hypertension 2025; 82:333-346. [PMID: 39624895 DOI: 10.1161/hypertensionaha.124.24151] [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: 10/11/2024] [Accepted: 11/13/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND Circulating proteins in blood are involved in various physiological processes, but their contributions to blood pressure regulation remain partially understood. In traditional observational studies, identifying circulating proteins causally associated with blood pressure is challenging because of potentially unmeasured confounding and possible reverse causality. METHODS Two-sample Mendelian randomization analyses were conducted to estimate the causal effects of 2270 circulating proteins (data sourced from 8 genome-wide association studies) on diastolic blood pressure, systolic blood pressure, and pulse pressure. Colocalization analyses were then used to investigate whether the circulating proteins and blood pressure traits shared causal genetic variants. To further verify the findings, we subsequently performed Steiger filtering analyses, annotation of protein-altering variants, assessment of overlap between protein quantitative trait loci and expression quantitative trait loci, protein-protein interaction and functional enrichment analyses, and drug target evaluation. To provide more potential biomarkers, we further evaluated the epidemiological associations of 2923 circulating proteins with blood pressure and hypertension by cross-sectional and longitudinal analyses using individual data in the UK Biobank. RESULTS Mendelian randomization and colocalization analyses identified 121 circulating proteins with putative causal effects on at least 1 blood pressure trait. Many of the identified proteins are enriched in the pathways relevant to blood pressure regulation, and a majority of these proteins are either known drug targets or druggable candidates. CONCLUSIONS This study has uncovered numerous circulating proteins potentially causally associated with blood pressure, providing insights into the regulatory mechanisms of blood pressure and potential therapeutic targets to facilitate blood pressure management.
Collapse
Affiliation(s)
- Siqi Xu
- Department of Rheumatology (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.), Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
- Clinical Research Centre, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.)
| | - Simin Wen
- Department of Rheumatology (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.), Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
- Clinical Research Centre, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.)
| | - Xizeng Zong
- Department of Rheumatology (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.), Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
- Clinical Research Centre, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.)
| | - Shifeng Wen
- Department of Rheumatology (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.), Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
- Clinical Research Centre, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.)
| | - Jianwei Zhu
- Department of Orthopedics (J.Z., W.Z.), Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Weipeng Zheng
- Department of Orthopedics (J.Z., W.Z.), Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhiqiang Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China (Z.W., P.C., C.D., Y.Z.)
| | - Peihua Cao
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China (Z.W., P.C., C.D., Y.Z.)
| | - Zhijiang Liang
- Department of Public Health, Guangdong Women and Children Hospital, Guangzhou, China (Z.L.)
| | - Changhai Ding
- Department of Rheumatology (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.), Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
- Clinical Research Centre, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.)
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China (Z.W., P.C., C.D., Y.Z.)
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia (C.D.)
| | - Yan Zhang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China (Z.W., P.C., C.D., Y.Z.)
| | - Guangfeng Ruan
- Department of Rheumatology (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.), Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
- Clinical Research Centre, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China (S.X., Simin Wen, X.Z., Shifeng Wen, C.D., G.R.)
| |
Collapse
|
2
|
Powell NR, Shugg T, Leighty J, Martin M, Kreutz RP, Eadon MT, Lai D, Lu T, Skaar TC. Analysis of the combined effect of rs699 and rs5051 on angiotensinogen expression and hypertension. Chronic Dis Transl Med 2024; 10:102-117. [PMID: 38872760 PMCID: PMC11166681 DOI: 10.1002/cdt3.103] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/23/2023] [Accepted: 11/06/2023] [Indexed: 06/15/2024] Open
Abstract
Background Hypertension (HTN) involves genetic variability in the renin-angiotensin system and influences antihypertensive response. We previously reported that angiotensinogen (AGT) messenger RNA (mRNA) is endogenously bound by miR-122-5p and rs699 A > G decreases reporter mRNA in the microRNA functional-assay PASSPORT-seq. The AGT promoter variant rs5051 C > T is in linkage disequilibrium (LD) with rs699 A > G and increases AGT transcription. The independent effect of these variants is understudied due to their LD therefore we aimed to test the hypothesis that increased AGT by rs5051 C > T counterbalances AGT decreased by rs699 A > G, and when these variants occur independently, it translates to HTN-related phenotypes. Methods We used in silico, in vitro, in vivo, and retrospective models to test this hypothesis. Results In silico, rs699 A > G is predicted to increase miR-122-5p binding affinity by 3%. Mir-eCLIP results show rs699 is 40-45 nucleotides from the strongest microRNA-binding site in the AGT mRNA. Unexpectedly, rs699 A > G increases AGT mRNA in an AGT-plasmid-cDNA HepG2 expression model. Genotype-Tissue Expression (GTEx) and UK Biobank analyses demonstrate liver AGT expression and HTN phenotypes are not different when rs699 A > G occurs independently from rs5051 C > T. However, GTEx and the in vitro experiments suggest rs699 A > G confers cell-type-specific effects on AGT mRNA abundance, and suggest paracrine renal renin-angiotensin-system perturbations could mediate the rs699 A > G associations with HTN. Conclusions We found that rs5051 C > T and rs699 A > G significantly associate with systolic blood pressure in Black participants in the UK Biobank, demonstrating a fourfold larger effect than in White participants. Further studies are warranted to determine if altered antihypertensive response in Black individuals might be due to rs5051 C > T or rs699 A > G. Studies like this will help clinicians move beyond the use of race as a surrogate for genotype.
Collapse
Affiliation(s)
- Nicholas R. Powell
- Division of Clinical Pharmacology, Department of MedicineSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Tyler Shugg
- Division of Clinical Pharmacology, Department of MedicineSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Jacob Leighty
- Division of Clinical Pharmacology, Department of MedicineSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Matthew Martin
- Department of Pharmacology and ToxicologySchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Rolf P. Kreutz
- Department of CardiologySchool of Medicine, Krannert Institute of Cardiology, Indiana UniversityIndianapolisIndianaUSA
| | - Michael T. Eadon
- Division of Nephrology, Department of MedicineSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Dongbing Lai
- Department of Medical and Molecular GeneticsSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Tao Lu
- Department of Pharmacology and ToxicologySchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Todd C. Skaar
- Division of Clinical Pharmacology, Department of MedicineSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| |
Collapse
|
3
|
Dmitrieva NI, Boehm M, Yancey PH, Enhörning S. Long-term health outcomes associated with hydration status. Nat Rev Nephrol 2024; 20:275-294. [PMID: 38409366 DOI: 10.1038/s41581-024-00817-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2024] [Indexed: 02/28/2024]
Abstract
Body water balance is determined by fundamental homeostatic mechanisms that maintain stable volume, osmolality and the composition of extracellular and intracellular fluids. Water balance is maintained by multiple mechanisms that continuously match water losses through urine, the skin, the gastrointestinal tract and respiration with water gains achieved through drinking, eating and metabolic water production. Hydration status is determined by the state of the water balance. Underhydration occurs when a decrease in body water availability, due to high losses or low gains, stimulates adaptive responses within the water balance network that are aimed at decreasing losses and increasing gains. This stimulation is also accompanied by cardiovascular adjustments. Epidemiological and experimental studies have linked markers of low fluid intake and underhydration - such as increased plasma concentration of vasopressin and sodium, as well as elevated urine osmolality - with an increased risk of new-onset chronic diseases, accelerated aging and premature mortality, suggesting that persistent activation of adaptive responses may be detrimental to long-term health outcomes. The causative nature of these associations is currently being tested in interventional trials. Understanding of the physiological responses to underhydration may help to identify possible mechanisms that underlie potential adverse, long-term effects of underhydration and inform future research to develop preventative and treatment approaches to the optimization of hydration status.
Collapse
Affiliation(s)
- Natalia I Dmitrieva
- Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, NIH, Bethesda, Maryland, USA.
| | - Manfred Boehm
- Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Paul H Yancey
- Biology Department, Whitman College, Walla Walla, Washington, USA
| | - Sofia Enhörning
- Perinatal and Cardiovascular Epidemiology, Lund University Diabetes Centre, Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
- Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
4
|
Daugherty A, Sawada H, Sheppard MB, Lu HS. Angiotensinogen as a Therapeutic Target for Cardiovascular and Metabolic Diseases. Arterioscler Thromb Vasc Biol 2024; 44:1021-1030. [PMID: 38572647 PMCID: PMC11225801 DOI: 10.1161/atvbaha.124.318374] [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] [Indexed: 04/05/2024]
Abstract
AGT (angiotensinogen) is the unique precursor for the generation of all the peptides of the renin-angiotensin system, but it has received relatively scant attention compared to many other renin-angiotensin system components. Focus on AGT has increased recently, particularly with the evolution of drugs to target the synthesis of the protein. AGT is a noninhibitory serpin that has several conserved domains in addition to the angiotensin II sequences at the N terminus. Increased study is needed on the structure-function relationship to resolve many unknowns regarding AGT metabolism. Constitutive whole-body genetic deletion of Agt in mice leads to multiple developmental defects creating a challenge to use these mice for mechanistic studies. This has been overcome by creating Agt-floxed mice to enable the development of cell-specific deficiencies that have provided considerable insight into a range of cardiovascular and associated diseases. This has been augmented by the recent development of pharmacological approaches targeting hepatocytes in humans to promote protracted inhibition of AGT synthesis. Genetic deletion or pharmacological inhibition of Agt has been demonstrated to be beneficial in a spectrum of diseases experimentally, including hypertension, atherosclerosis, aortic and superior mesenteric artery aneurysms, myocardial dysfunction, and hepatic steatosis. This review summarizes the findings of recent studies utilizing AGT manipulation as a therapeutic approach.
Collapse
Affiliation(s)
- Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Mary B. Sheppard
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
- Department of Family and Community Medicine, University of Kentucky, Lexington, KY
- Department of Surgery, University of Kentucky, Lexington, KY
| | - Hong S. Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
5
|
Schiffrin EL. RNA Injection Every 6 Months to Improve Adherence and Lower Blood Pressure in Patients With Hypertension. JAMA 2024; 331:733-735. [PMID: 38363578 DOI: 10.1001/jama.2023.26071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Affiliation(s)
- Ernesto L Schiffrin
- Lady Davis Institute for Medical Research, and Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montréal, Québec, Canada
| |
Collapse
|
6
|
Rodrigues AF, Bader M. The contribution of the AT1 receptor to erythropoiesis. Biochem Pharmacol 2023; 217:115805. [PMID: 37714274 DOI: 10.1016/j.bcp.2023.115805] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
The renin-angiotensin system (RAS) comprises a broad set of functional peptides and receptors that play a role in cardiovascular homeostasis and contribute to cardiovascular pathologies. Angiotensin II (Ang II) is the most potent peptide hormone produced by the RAS due to its high abundance and its strong and pleiotropic impact on the cardiovascular system. Formation of Ang II takes place in the bloodstream and additionally in tissues in the so-called local RAS. Of the two Ang II receptors (AT1 and AT2) that Ang II binds to, AT1 is the most expressed throughout the mammalian body. AT1 expression is not restricted to cells of the cardiovascular system but in fact AT1 protein is found in nearly all organs, hence, Ang II takes part in several modulatory physiological processes one of which is erythropoiesis. In this review, we present multiple evidence supporting that Ang II modulates physiological and pathological erythropoiesis processes trough the AT1 receptor. Cumulative evidence indicates that Ang II by three distinct mechanisms influences erythropoiesis: 1) stimulation of renal erythropoietin synthesis; 2) direct action on bone marrow precursor cells; and 3) modulation of sympathetic nerve activity to the bone marrow. The text highlights clinical and preclinical evidence focusing on mechanistic studies using rodent models.
Collapse
Affiliation(s)
- André F Rodrigues
- Max Delbrück Center (MDC), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany.
| | - Michael Bader
- Max Delbrück Center (MDC), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany; Charité Universitätsmedizin Berlin, Berlin, Germany; Institute for Biology, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
7
|
Peltekian L, Gasparini S, Fazan FS, Karthik S, Iverson G, Resch JM, Geerling JC. Sodium appetite and thirst do not require angiotensinogen production in astrocytes or hepatocytes. J Physiol 2023; 601:3499-3532. [PMID: 37291801 DOI: 10.1113/jp283169] [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: 12/28/2022] [Accepted: 06/02/2023] [Indexed: 06/10/2023] Open
Abstract
In addition to its renal and cardiovascular functions, angiotensin signalling is thought to be responsible for the increases in salt and water intake caused by hypovolaemia. However, it remains unclear whether these behaviours require angiotensin production in the brain or liver. Here, we use in situ hybridization to identify tissue-specific expression of the genes required for producing angiotensin peptides, and then use conditional genetic deletion of the angiotensinogen gene (Agt) to test whether production in the brain or liver is necessary for sodium appetite and thirst. In the mouse brain, we identified expression of Agt (the precursor for all angiotensin peptides) in a large subset of astrocytes. We also identified Ren1 and Ace (encoding enzymes required to produce angiotensin II) expression in the choroid plexus, and Ren1 expression in neurons within the nucleus ambiguus compact formation. In the liver, we confirmed that Agt is widely expressed in hepatocytes. We next tested whether thirst and sodium appetite require angiotensinogen production in astrocytes or hepatocytes. Despite virtually eliminating expression in the brain, deleting astrocytic Agt did not reduce thirst or sodium appetite. Despite markedly reducing angiotensinogen in the blood, eliminating Agt from hepatocytes did not reduce thirst or sodium appetite, and in fact, these mice consumed the largest amounts of salt and water after sodium deprivation. Deleting Agt from both astrocytes and hepatocytes also did not prevent thirst or sodium appetite. Our findings suggest that angiotensin signalling is not required for sodium appetite or thirst and highlight the need to identify alternative signalling mechanisms. KEY POINTS: Angiotensin signalling is thought to be responsible for the increased thirst and sodium appetite caused by hypovolaemia, producing elevated water and sodium intake. Specific cells in separate brain regions express the three genes needed to produce angiotensin peptides, but brain-specific deletion of the angiotensinogen gene (Agt), which encodes the lone precursor for all angiotensin peptides, did not reduce thirst or sodium appetite. Double-deletion of Agt from brain and liver also did not reduce thirst or sodium appetite. Liver-specific deletion of Agt reduced circulating angiotensinogen levels without reducing thirst or sodium appetite. Instead, these angiotensin-deficient mice exhibited an enhanced sodium appetite. Because the physiological mechanisms controlling thirst and sodium appetite continued functioning without angiotensin production in the brain and liver, understanding these mechanisms requires a renewed search for the hypovolaemic signals necessary for activating each behaviour.
Collapse
Affiliation(s)
- Lila Peltekian
- Department of Neurology, University of Iowa, Iowa City, IA, USA
| | | | | | | | | | - Jon M Resch
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Joel C Geerling
- Department of Neurology, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
8
|
Youn EK, Cho HM, Jung JK, Yoon GE, Eto M, Kim JI. Pathologic HDAC1/c-Myc signaling axis is responsible for angiotensinogen transcription and hypertension induced by high-fat diet. Biomed Pharmacother 2023; 164:114926. [PMID: 37244179 DOI: 10.1016/j.biopha.2023.114926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/03/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023] Open
Abstract
High-fat diet (HFD)-induced obesity is a cause of resistant hypertension. We have shown a possible link between histone deacetylases (HDACs) and renal angiotensinogen (Agt) upregulation in the HFD-induced hypertension, whereas the underlying mechanisms remain to be elucidated. Here, using a HDAC1/2 inhibitor romidepsin (FK228) and siRNAs, we determined roles of HDAC1 and HDAC2 in HFD-induced hypertension and found the pathologic signaling axis between HDAC1 and Agt transcription. Treatment with FK228 canceled the increased blood pressure of male C57BL/6 mice induced by HFD. FK228 also blocked upregulation of renal Agt mRNA, protein, angiotensin II (Ang II) or serum Ang II. Activation and nuclear accumulation of both HDAC1 and HDAC2 occurred in the HFD group. The HFD-induced HDAC activation was associated with an increase in deacetylated c-Myc transcription factor. Silencing of HDAC1, HDAC2 or c-Myc in HRPTEpi cells decreased Agt expression. However, only HDAC1 knockdown, but not HDAC2, increased c-Myc acetylation, suggesting selective roles in two enzymes. Chromatin immunoprecipitation assay revealed that HFD induced the binding of HDAC1 and deacetylated c-Myc at the Agt gene promoter. A putative c-Myc binding sequence in the promotor region was necessary for Agt transcription. Inhibition of c-Myc downregulated Agt and Ang II levels in kidney and serum, ameliorating HFD-induced hypertension. Thus, the abnormal HDAC1/2 in the kidney may be responsible for the upregulation of the Agt gene expression and hypertension. The results expose the pathologic HDAC1/c-myc signaling axis in kidney as a promising therapeutic target for obesity-associated resistant hypertension.
Collapse
Affiliation(s)
- Eui Kyung Youn
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Hyun Min Cho
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Jin Ki Jung
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Ga-Eun Yoon
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Masumi Eto
- Department of Veterinary Medicine, Okayama University of Science, Ehime 794-8555, Japan
| | - Jee In Kim
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu 42601, Republic of Korea.
| |
Collapse
|
9
|
Powell NR, Shugg T, Leighty J, Martin M, Kreutz RP, Eadon MT, Lai D, Lu T, Skaar TC. Analysis of the Combined Effect of rs699 and rs5051 on Angiotensinogen Expression and Hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.07.536073. [PMID: 37066278 PMCID: PMC10104131 DOI: 10.1101/2023.04.07.536073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Hypertension (HTN) involves genetic variability in the renin-angiotensin system and characterizing this variability will help advance precision antihypertensive treatments. We previously reported that angiotensinogen (AGT) mRNA is endogenously bound by mir-122-5p and that rs699 A>G significantly decreases reporter mRNA in the functional mirSNP assay PASSPORT-seq. The AGT promoter variant rs5051 C>T is in linkage disequilibrium (LD) with rs699 A>G and increases AGT transcription. We hypothesized that the increased AGT by rs5051 C>T counterbalances AGT decrease by rs699 A>G, and when these variants occur independently, would translate to HTN-related phenotypes. The independent effect of each of these variants is understudied due to their LD, therefore, we used in silico, in vitro, in vivo, and retrospective clinical and biobank analyses to assess HTN and AGT expression phenotypes where rs699 A>G occurs independently from rs5051 C>T. In silico, rs699 A>G is predicted to increase mir-122-5p binding strength by 3%. Mir-eCLIP assay results show that rs699 is 40-45 nucleotides from the strongest microRNA binding site in the AGT mRNA. Unexpectedly, rs699 A>G increases AGT mRNA in a plasmid cDNA HepG2 expression model. GTEx and UK Biobank analyses demonstrate that liver AGT expression and HTN phenotypes were not different when rs699 A>G occurs independently from rs5051 C>T, allowing us to reject the original hypothesis. However, both GTEx and our in vitro experiments suggest rs699 A>G confers cell-type specific effects on AGT mRNA abundance. We found that rs5051 C>T and rs699 A>G significantly associate with systolic blood pressure in Black participants in the UK Biobank, demonstrating a 4-fold larger effect than in White participants. Further studies are warranted to determine if the altered antihypertensive response in Black individuals might be due to rs5051 C>T or rs699 A>G. Studies like this will help clinicians move beyond the use of race as a surrogate for genotype.
Collapse
Affiliation(s)
- Nicholas R. Powell
- Indiana University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Indianapolis IN
| | - Tyler Shugg
- Indiana University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Indianapolis IN
| | - Jacob Leighty
- Indiana University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Indianapolis IN
| | - Matthew Martin
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, Indianapolis IN
| | - Rolf P. Kreutz
- Indiana University School of Medicine, Department of Cardiology, Krannert Institute of Cardiology, Indianapolis IN
| | - Michael T. Eadon
- Indiana University School of Medicine, Department of Medicine, Division of Nephrology, Indianapolis IN
- Indiana University School of Medicine, Department of Medical and Molecular Genetics, Indianapolis IN
| | - Dongbing Lai
- Indiana University School of Medicine, Department of Medical and Molecular Genetics, Indianapolis IN
| | - Tao Lu
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, Indianapolis IN
| | - Todd C. Skaar
- Indiana University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Indianapolis IN
- Indiana University School of Medicine, Department of Medical and Molecular Genetics, Indianapolis IN
| |
Collapse
|
10
|
Yamashita K, Morimoto S, Inoue Y, Hirata K, Kimura S, Seki Y, Bokuda K, Watanabe D, Ichihara A. A case of a pregnant woman with primary aldosteronism and superimposed preeclampsia treated with esaxerenone. J Endocr Soc 2022; 6:bvac085. [PMID: 35733831 PMCID: PMC9206722 DOI: 10.1210/jendso/bvac085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Indexed: 11/19/2022] Open
Abstract
Abstract
During pregnancy, there is no established treatment for idiopathic hyperaldosteronism (IHA), the most common form of primary aldosteronism (PA) due to bilateral adrenal hyperplasia. Here, we report the case of a pregnant patient with IHA who was successfully treated with esaxerenone, a non-steroidal mineralocorticoid receptor (MR) antagonist. A 39-year-old woman, was diagnosed with IHA and commenced on nifedipine 20 mg daily because she desired to be pregnant. After one year, she became pregnant. Her blood pressure was well controlled until 34 weeks of gestation when her home blood pressure became elevated up to 140/90 mmHg. Although the dose of nifedipine was increased to 80 mg daily, her blood pressure increased to 151/97 mmHg and urinary test showed proteinuria of 2+ in 35 weeks of gestation. She was diagnosed with superimposed preeclampsia (SPE) and additionally treated with esaxerenone. Her blood pressure decreased to 120-140/98-100 mmHg and the proteinuria improved to +/-. A successful cesarean section at 37 weeks resulted in the delivery of a healthy baby boy. Her blood pressure was well controlled although esaxerenone was discontinued 2 weeks after the delivery. This is the first case of a pregnant woman who was safely treated with esaxerenone despite being a female at advanced maternal age who had been diagnosed with IHA and developed SPE. Further studies are needed to investigate the efficacy and safety of non-steroidal selective MR antagonist in similar pregnant patients with IHA, to establish better treatment strategy for these patients.
Collapse
Affiliation(s)
- Kaoru Yamashita
- Department of Endocrinology and Hypertension, Tokyo Women’s Medical University, 8-1 Kawada-cho , Shinjuku-ku, Tokyo 162-8666, Japan
| | - Satoshi Morimoto
- Department of Endocrinology and Hypertension, Tokyo Women’s Medical University, 8-1 Kawada-cho , Shinjuku-ku, Tokyo 162-8666, Japan
| | - Yuko Inoue
- Department of Endocrinology and Hypertension, Tokyo Women’s Medical University, 8-1 Kawada-cho , Shinjuku-ku, Tokyo 162-8666, Japan
| | - Kiyotaka Hirata
- Department of Endocrinology and Hypertension, Tokyo Women’s Medical University, 8-1 Kawada-cho , Shinjuku-ku, Tokyo 162-8666, Japan
| | - Shihori Kimura
- Department of Endocrinology and Hypertension, Tokyo Women’s Medical University, 8-1 Kawada-cho , Shinjuku-ku, Tokyo 162-8666, Japan
| | - Yasufumi Seki
- Department of Endocrinology and Hypertension, Tokyo Women’s Medical University, 8-1 Kawada-cho , Shinjuku-ku, Tokyo 162-8666, Japan
| | - Kanako Bokuda
- Department of Endocrinology and Hypertension, Tokyo Women’s Medical University, 8-1 Kawada-cho , Shinjuku-ku, Tokyo 162-8666, Japan
| | - Daisuke Watanabe
- Department of Endocrinology and Hypertension, Tokyo Women’s Medical University, 8-1 Kawada-cho , Shinjuku-ku, Tokyo 162-8666, Japan
| | - Atsuhiro Ichihara
- Department of Endocrinology and Hypertension, Tokyo Women’s Medical University, 8-1 Kawada-cho , Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
11
|
Abstract
BACKGROUND CACNA1D gene, which encodes the α1 subunit of the Cav1.3 L-type calcium channel effectively regulates intracellular Ca2+ stability. In recent years, clinical studies have shown that the CACNA1D polymorphisms were associated with hypertension. OBJECTIVE The purpose of this study was to evaluate the effects of CACNA1D exon mutation on blood pressure (BP) in Sprague-Dawley rats. METHODS The rats with CACNA1D p.D307G, CACNA1D p.V936I or CACNA1D p.R1516Q were constructed using CRISPR-Cas9 technology. SBP measurements of rats were taken for 32 weeks. Tissue morphology of rats and vasoactive substances in serum was tested. Furthermore, the effects of L-type calcium channel blocker isradipine and endothelin-1 (ET-1) inhibitor BQ-123 on BP of double mutation rats (CACNA1D p.D307G/p.R1516Q) were tested. Then we examined the effects of CACNA1D gene mutation on gene expression in human umbilical vein endothelial cells (HUVECs) and vascular smooth muscle cells (VSMCs). RESULTS Elevated SBP and increased circulating ET-1 was observed in CACNA1D p.D307G mutant rats. Morphological assessments showed that the vascular, cardiac and renal remodeling could also be observed in rats with p.D307G mutant. Cav1.3 protein expression and calcineurin phosphatase activity in VSMCs of rats with CACNA1D p.D307G were increased in vitro, and the vascular ring tension test of mesenteric grade 3 arteries in CACNA1D p.D307G rats were increased in vivo. Furthermore, ET-1 expression were increased in isolated primary aortic endothelial cells in p.D307G mutant rats and transfected p.D307G mutant HUVECs. Finally, double heterozygosity rats with CACNA1D p.D307G/p.R1516Q or CACNA1D p.D307G/p.V936I further accelerated the rise of SBP compared with p.D307G mutation rats, and isradipine and BQ-123 reduced BP to the same extent in CACNA1D p.D307G/p.R1516Q rats. CONCLUSION CACNA1D gene is key players in the regulation of blood pressure. CACNA1D mutation rat may be a new hypertension animal model.
Collapse
|
12
|
Habeeb E, Aldosari S, Saghir SA, Cheema M, Momenah T, Husain K, Omidi Y, Rizvi SA, Akram M, Ansari RA. Role of environmental toxicants in the development of hypertensive and cardiovascular diseases. Toxicol Rep 2022; 9:521-533. [PMID: 35371924 PMCID: PMC8971584 DOI: 10.1016/j.toxrep.2022.03.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
The incidence of hypertension with diabetes mellitus (DM) as a co-morbid condition is on the rise worldwide. In 2000, an estimated 972 million adults had hypertension, which is predicted to grow to 1.56 billion by 2025. Hypertension often leads to diabetes mellitus that strongly puts the patients at an increased risk of cardiovascular, kidney, and/or atherosclerotic diseases. Hypertension has been identified as a major risk factor for the development of diabetes; patients with hypertension are at two-to-three-fold higher risk of developing diabetes than patients with normal blood pressure (BP). Causes for the increase in hypertension and diabetes are not well understood, environmental factors (e.g., exposure to environmental toxicants like heavy metals, organic solvents, pesticides, alcohol, and urban lifestyle) have been postulated as one of the reasons contributing to hypertension and cardiovascular diseases (CVD). The mechanism of action(s) of these toxicants in developing hypertension and CVDs is not well defined. Research studies have linked hypertension with the chronic consumption of alcohol and exposure to metals like lead, mercury, and arsenic have also been linked to hypertension and CVD. Workers chronically exposed to styrene have a higher incidence of CVD. Recent studies have demonstrated that exposure to particulate matter (PM) in diesel exhaust and urban air contributes to increased CVD and mortality. In this review, we have imparted the role of environmental toxicants such as heavy metals, organic pollutants, PM, alcohol, and some drugs in hypertension and CVD along with possible mechanisms and limitations in extrapolating animal data to humans.
Collapse
Affiliation(s)
- Ehsan Habeeb
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200S University Drive, Fort Lauderdale, FL 33200, USA
| | - Saad Aldosari
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200S University Drive, Fort Lauderdale, FL 33200, USA
| | - Shakil A. Saghir
- The Scotts Company LLC, Marysville, OH 43041, USA
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Mariam Cheema
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200S University Drive, Fort Lauderdale, FL 33200, USA
| | - Tahani Momenah
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200S University Drive, Fort Lauderdale, FL 33200, USA
| | - Kazim Husain
- Department of Gastrointestinal Oncology (FOB-2), Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200S University Drive, Fort Lauderdale, FL 33200, USA
| | - Syed A.A. Rizvi
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, VA 23668, USA
| | - Muhammad Akram
- Department of Eastern Medicine and Surgery, Government College University Faisalabad, Faisalabad, Pakistan
| | - Rais A. Ansari
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200S University Drive, Fort Lauderdale, FL 33200, USA
| |
Collapse
|
13
|
Xu C, Liu C, Xiong J, Yu J. Cardiovascular aspects of the (pro)renin receptor: Function and significance. FASEB J 2022; 36:e22237. [PMID: 35226776 DOI: 10.1096/fj.202101649rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/13/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases (CVDs), including all types of disorders related to the heart or blood vessels, are the major public health problems and the leading causes of mortality globally. (Pro)renin receptor (PRR), a single transmembrane protein, is present in cardiomyocytes, vascular smooth muscle cells, and endothelial cells. PRR plays an essential role in cardiovascular homeostasis by regulating the renin-angiotensin system and several intracellular signals such as mitogen-activated protein kinase signaling and wnt/β-catenin signaling in various cardiovascular cells. This review discusses the current evidence for the pathophysiological roles of the cardiac and vascular PRR. Activation of PRR in cardiomyocytes may contribute to myocardial ischemia/reperfusion injury, cardiac hypertrophy, diabetic or alcoholic cardiomyopathy, salt-induced heart damage, and heart failure. Activation of PRR promotes vascular smooth muscle cell proliferation, endothelial cell dysfunction, neovascularization, and the progress of vascular diseases. In addition, phenotypes of animals transgenic for PRR and the hypertensive actions of PRR in the brain and kidney and the soluble PRR are also discussed. Targeting PRR in local tissues may offer benefits for patients with CVDs, including heart injury, atherosclerosis, and hypertension.
Collapse
Affiliation(s)
- Chuanming Xu
- Translational Medicine Centre, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Chunju Liu
- Department of Clinical Laboratory, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Jianhua Xiong
- Department of Cardiology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Jun Yu
- Center for Metabolic Disease Research and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Herrera P, Cauchi RJ. ACE and ACE2: insights from Drosophila and implications for COVID-19. Heliyon 2021; 7:e08555. [PMID: 34901515 PMCID: PMC8648576 DOI: 10.1016/j.heliyon.2021.e08555] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/21/2021] [Accepted: 12/02/2021] [Indexed: 12/27/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) and its homologue ACE2 are key regulators of the renin-angiotensin system and thereby cardiovascular function through their zinc-metallopeptidase activity on vasoactive peptides. ACE2 also serves as the receptor for the cellular entry of various coronaviruses including the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is responsible for the coronavirus disease 2019 (COVID-19). The unprecedented scale of the COVID-19 pandemic has spurred the use of mammalian models to investigate the SARS-ACE2 relationship and knowledge gained from such research has accelerated development of vaccines and therapeutics. Recent studies have just started to underscore the utility of the fruit fly Drosophila melanogaster as a model system to study virus-host interactions and pathogenicity. Notably, the remarkable existence of catalytically functional ACE and ACE2 orthologues in Drosophila, discovered more than two decades ago, provides a unique opportunity for further developing this model organism to better understand COVID-19 in addition to identifying coronavirus preventative and therapeutic interventions targeting ACE2. Here, we review the studies that revealed crucial insights on the biochemistry and physiology of Ance and Acer, two out of the six Drosophila ACE family members with the greatest homology to human ACE and ACE2. We highlight shared in vivo functions outside of the renin-angiotensin system, which is not conserved in flies. Importantly, we identify knowledge gaps that can be filled by further research and outline ways that can raise Drosophila to a powerful model system to combat SARS-CoV-2 and its threatening vaccine-evading variants.
Collapse
Affiliation(s)
- Paul Herrera
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences Building, University of Malta, Msida, Malta
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Ruben J. Cauchi
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences Building, University of Malta, Msida, Malta
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Corresponding author.
| |
Collapse
|
15
|
Abstract
Chronic cardiovascular diseases are associated with inflammatory responses within the blood vessels and end organs. The origin of this inflammation has not been certain, and neither is its relationship to disease clear. There is a need to determine whether this association is causal or coincidental to the processes leading to cardiovascular disease. These processes are themselves complex: many cardiovascular diseases arise in conjunction with the presence of sustained elevation of blood pressure. Inflammatory processes have been linked to hypertension, and causality has been suggested. Evidence of causality poses the difficult challenge of linking the integrated and multifaceted biology of blood pressure regulation with vascular function and complex elements of immune system function. These include both, innate and adaptive immunity, as well as interactions between the host immune system and the omnipresent microorganisms that are encountered in the environment and that colonize and exist in commensal relationship with the host. Progress has been made in this task and has drawn on experimental approaches in animals, much of which have focused on hypertension occurring with prolonged infusion of angiotensin II. These laboratory studies are complemented by studies that seek to inform disease mechanism by examining the genomic basis of heritable disease susceptibility in human populations. In this realm too, evidence has emerged that implicates genetic variation affecting immunity in disease pathogenesis. In this article, we survey the genetic and genomic evidence linking high blood pressure and its end-organ injuries to immune system function and examine evidence that genomic factors can influence disease risk. © 2021 American Physiological Society. Compr Physiol 11:1-22, 2021.
Collapse
Affiliation(s)
- Isha S Dhande
- Center for Human Genetics, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Peter A Doris
- Center for Human Genetics, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
16
|
Sheng X, Guan Y, Ma Z, Wu J, Liu H, Qiu C, Vitale S, Miao Z, Seasock MJ, Palmer M, Shin MK, Duffin KL, Pullen SS, Edwards TL, Hellwege JN, Hung AM, Li M, Voight BF, Coffman TM, Brown CD, Susztak K. Mapping the genetic architecture of human traits to cell types in the kidney identifies mechanisms of disease and potential treatments. Nat Genet 2021; 53:1322-1333. [PMID: 34385711 PMCID: PMC9338440 DOI: 10.1038/s41588-021-00909-9] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 06/30/2021] [Indexed: 02/07/2023]
Abstract
The functional interpretation of genome-wide association studies (GWAS) is challenging due to the cell-type-dependent influences of genetic variants. Here, we generated comprehensive maps of expression quantitative trait loci (eQTLs) for 659 microdissected human kidney samples and identified cell-type-eQTLs by mapping interactions between cell type abundances and genotypes. By partitioning heritability using stratified linkage disequilibrium score regression to integrate GWAS with single-cell RNA sequencing and single-nucleus assay for transposase-accessible chromatin with high-throughput sequencing data, we prioritized proximal tubules for kidney function and endothelial cells and distal tubule segments for blood pressure pathogenesis. Bayesian colocalization analysis nominated more than 200 genes for kidney function and hypertension. Our study clarifies the mechanism of commonly used antihypertensive and renal-protective drugs and identifies drug repurposing opportunities for kidney disease.
Collapse
Affiliation(s)
- Xin Sheng
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuting Guan
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ziyuan Ma
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Junnan Wu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongbo Liu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Chengxiang Qiu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Vitale
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhen Miao
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew J Seasock
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Palmer
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Kevin L Duffin
- Eli Lilly and Company Lilly Corporate Center, Indianapolis, IN, USA
| | - Steven S Pullen
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jacklyn N Hellwege
- Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adriana M Hung
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mingyao Li
- Department of Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Benjamin F Voight
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas M Coffman
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | | | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA.
- Institute of Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Emathinger JM, Nelson JW, Gurley SB. Advances in use of mouse models to study the renin-angiotensin system. Mol Cell Endocrinol 2021; 529:111255. [PMID: 33789143 PMCID: PMC9119406 DOI: 10.1016/j.mce.2021.111255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/19/2021] [Accepted: 03/20/2021] [Indexed: 12/28/2022]
Abstract
The renin-angiotensin system (RAS) is a highly complex hormonal cascade that spans multiple organs and cell types to regulate solute and fluid balance along with cardiovascular function. Much of our current understanding of the functions of the RAS has emerged from a series of key studies in genetically-modified animals. Here, we review key findings from ground-breaking transgenic models, spanning decades of research into the RAS, with a focus on their use in studying blood pressure. We review the physiological importance of this regulatory system as evident through the examination of mouse models for several major RAS components: angiotensinogen, renin, ACE, ACE2, and the type 1 A angiotensin receptor. Both whole-animal and cell-specific knockout models have permitted critical RAS functions to be defined and demonstrate how redundancy and multiplicity within the RAS allow for compensatory adjustments to maintain homeostasis. Moreover, these models present exciting opportunities for continued discovery surrounding the role of the RAS in disease pathogenesis and treatment for cardiovascular disease and beyond.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2/deficiency
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensinogen/deficiency
- Angiotensinogen/genetics
- Animals
- Blood Pressure/genetics
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Disease Models, Animal
- Gene Expression Regulation
- Humans
- Kidney/cytology
- Kidney/metabolism
- Mice
- Mice, Knockout
- Receptor, Angiotensin, Type 1/deficiency
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 2/deficiency
- Receptor, Angiotensin, Type 2/genetics
- Renin/deficiency
- Renin/genetics
- Renin-Angiotensin System/genetics
- Signal Transduction
- Water-Electrolyte Balance/genetics
Collapse
Affiliation(s)
- Jacqueline M Emathinger
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR, USA.
| | - Jonathan W Nelson
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR, USA.
| | - Susan B Gurley
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
18
|
Chen Z, Huo X, Chen G, Luo X, Xu X. Lead (Pb) exposure and heart failure risk. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:28833-28847. [PMID: 33840028 DOI: 10.1007/s11356-021-13725-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/25/2021] [Indexed: 02/08/2023]
Abstract
Lead (Pb) is a heavy metal with widespread industrial use, but it is also a widespread environmental contaminant with serious toxicological consequences to many species. Pb exposure adversely impacts the cardiovascular system in humans, leading to cardiac dysfunction, but its effects on heart failure risk remain poorly elucidated. To better understand the pathophysiological effects of Pb, we review potential mechanisms by which Pb exposure leads to cardiac dysfunction. Adverse effects of Pb exposure on cardiac function include heart failure risk, pressure overload, arrhythmia, myocardial ischemia, and cardiotoxicity. The data reviewed clearly establish that Pb exposure can play an important role in the occurrence and development of heart failure. Future epidemiological and mechanistic studies should be developed to better understand the involvement of Pb exposure in heart failure.
Collapse
Affiliation(s)
- Zihan Chen
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, 22 Xinling Rd, Shantou, 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China
| | - Guangcan Chen
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, 22 Xinling Rd, Shantou, 515041, Guangdong, China
| | - Xiuli Luo
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, 22 Xinling Rd, Shantou, 515041, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, 22 Xinling Rd, Shantou, 515041, Guangdong, China.
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
19
|
Shu Z, Wan J, Read RJ, Carrell RW, Zhou A. Angiotensinogen and the Modulation of Blood Pressure. Front Cardiovasc Med 2021; 8:645123. [PMID: 33816576 PMCID: PMC8012498 DOI: 10.3389/fcvm.2021.645123] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
The angiotensin peptides that control blood pressure are released from the non-inhibitory plasma serpin, angiotensinogen, on cleavage of its extended N-terminal tail by the specific aspartyl-protease, renin. Angiotensinogen had previously been assumed to be a passive substrate, but we describe here how recent studies reveal an inherent conformational mechanism that is critical to the cleavage and release of the angiotensin peptides and consequently to the control of blood pressure. A series of crystallographic structures of angiotensinogen and its derivative forms, together with its complexes with renin show in molecular detail how the interaction with renin triggers a profound shift of the amino-terminal tail of angiotensinogen with modulation occurring at several levels. The tail of angiotensinogen is restrained by a labile disulfide bond, with changes in its redox status affecting angiotensin release, as demonstrably so in the hypertensive complication of pregnancy, pre-eclampsia. The shift of the tail also enhances the binding of renin through a tail-in-mouth allosteric mechanism. The N-terminus is now seen to insert into a pocket equivalent to the hormone-binding site on other serpins, with helix H of angiotensinogen unwinding to form key interactions with renin. The findings explain the precise species specificity of the interaction with renin and with variant carbohydrate linkages. Overall, the studies provide new insights into the physiological regulation of angiotensin release, with an ability to respond to local tissue and temperature changes, and with the opening of strategies for the development of novel agents for the treatment of hypertension.
Collapse
Affiliation(s)
- Zimei Shu
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahui Wan
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Randy J Read
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Robin W Carrell
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Aiwu Zhou
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Saadi S, Ghazali HM, Saari N, Abdulkarim SM. The structural reconformation of peptides in enhancing functional and therapeutic properties: Insights into their solid state crystallizations. Biophys Chem 2021; 273:106565. [PMID: 33780688 DOI: 10.1016/j.bpc.2021.106565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 01/05/2023]
Abstract
Therapeutic peptides derived proteins with alpha-reconformation states like antibody shape have shown potential effects in combating terrible diseases linked with earlier signs of angiogensis, mutagenesis and transgenesis. Alpha reconformation in material design refers to the folding of the peptide chains and their transitions under reversible chemical bonds of disulfide chemical bridges and further non-covalence lesions. Thus, the rational design of signal peptides into alpha-helix is intended in increasing the defending effects of peptides into cores like adjuvant antibiotic and/or vaccines. Thereby, the signal peptides are able in displaying multiple eradicating regions by changing crystal-depositions and deviation angles. These types of molecular structures could have multiple advantages in tracing disease syndromes and impurities by increasing the host defense against the fates of pathogens and viruses, eventually leading to the loss in signaling by increasing peptide susceptibility levels to folding and unfolding and therefore, formation of transgenic peptide models. Alpha reconformation peptides is aimed in triggering as well as other regulatory functions such as remodulating metabolic chain disorders of lipolysis and glucolysis by increasing the insulin and leptin resistance for best lipid storages and lipoprotein density distributions.
Collapse
Affiliation(s)
- Sami Saadi
- Institut de la Nutrition, de l'Alimentation et des Technologies Agro-alimentaires INATAA 25017, Université Frères Mentouri, Constantine 1, Algeria; Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Hasanah Mohd Ghazali
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Nazamid Saari
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Sabo Mohammed Abdulkarim
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
21
|
Hebert JF, Millar JA, Raghavan R, Romney A, Podrabsky JE, Rennie MY, Felker AM, O'Tierney-Ginn P, Morita M, DuPriest EA, Morgan TK. Male fetal sex affects uteroplacental angiogenesis in growth restriction mouse model†. Biol Reprod 2021; 104:924-934. [PMID: 33459759 DOI: 10.1093/biolre/ioab006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/11/2020] [Accepted: 01/13/2021] [Indexed: 01/28/2023] Open
Abstract
Abnormally increased angiotensin II activity related to maternal angiotensinogen (AGT) genetic variants, or aberrant receptor activation, is associated with small-for-gestational-age babies and abnormal uterine spiral artery remodeling in humans. Our group studies a murine AGT gene titration transgenic (TG; 3-copies of the AGT gene) model, which has a 20% increase in AGT expression mimicking a common human AGT genetic variant (A[-6]G) associated with intrauterine growth restriction (IUGR) and spiral artery pathology. We hypothesized that aberrant maternal AGT expression impacts pregnancy-induced uterine spiral artery angiogenesis in this mouse model leading to IUGR. We controlled for fetal sex and fetal genotype (e.g., only 2-copy wild-type [WT] progeny from WT and TG dams were included). Uteroplacental samples from WT and TG dams from early (days 6.5 and 8.5), mid (d12.5), and late (d16.5) gestation were studied to assess uterine natural killer (uNK) cell phenotypes, decidual metrial triangle angiogenic factors, placental growth and capillary density, placental transcriptomics, and placental nutrient transport. Spiral artery architecture was evaluated at day 16.5 by contrast-perfused three-dimensional microcomputed tomography (3D microCT). Our results suggest that uteroplacental angiogenesis is significantly reduced in TG dams at day 16.5. Males from TG dams are associated with significantly reduced uteroplacental angiogenesis from early to late gestation compared with their female littermates and WT controls. Angiogenesis was not different between fetal sexes from WT dams. We conclude that male fetal sex compounds the pathologic impact of maternal genotype in this mouse model of growth restriction.
Collapse
Affiliation(s)
- Jessica F Hebert
- Department of Pathology, Oregon Health & Science University, Portland, OR, USA.,Center for Developmental Health, Oregon Health & Science University, Portland, OR, USA.,Department of Biology, Portland State University, Portland, OR, USA
| | - Jess A Millar
- Department of Biology, Portland State University, Portland, OR, USA
| | - Rahul Raghavan
- Department of Biology, Portland State University, Portland, OR, USA
| | - Amie Romney
- Department of Biology, Portland State University, Portland, OR, USA
| | | | - Monique Y Rennie
- Center for Developmental Health, Oregon Health & Science University, Portland, OR, USA
| | - Allison M Felker
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | | | - Mayu Morita
- Department of Pathology, Oregon Health & Science University, Portland, OR, USA.,Center for Developmental Health, Oregon Health & Science University, Portland, OR, USA
| | - Elizabeth A DuPriest
- Center for Developmental Health, Oregon Health & Science University, Portland, OR, USA.,Division of Natural Sciences and Health, Warner Pacific University, Portland, OR, USA
| | - Terry K Morgan
- Department of Pathology, Oregon Health & Science University, Portland, OR, USA.,Center for Developmental Health, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
22
|
The effects of sacubitril/valsartan and ramipril on the male fertility in hypertensive rats. North Clin Istanb 2020; 7:425-432. [PMID: 33163876 PMCID: PMC7603857 DOI: 10.14744/nci.2020.30906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 05/29/2020] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE: Renin angiotensinogen system (RAS) inhibitors, ramipril and sacubitril/valsartan are frequently used in the treatment of cardiovascular diseases. Although they are known as contraindicated during pregnancy in hypertensive women, there is not any outcome of their safety in male fertility after exposure to ramipril or sacubitril/valsartan. In this study, we aimed to evaluate the effects of ramipril and sacubitril/valsartan to highlight their safety in the male fertility in normotensive and hypertensive rats. METHODS: Adult male normotensive and dexamethasone-induced hypertensive rats were treated with sacubitril/valsartan, ramipril and saline for 18 days. Arterial blood pressures were verified using carotid artery cannulation. Male fertility parameters, including the testis weights, histopathologic scoring of the testis, sperm count, sperm motility, morphology, and serum testosterone levels, were analyzed in treated and nontreated normotensive/hypertensive rats. RESULTS: Sacubitril/valsartan or ramipril treatments did not reveal a significant difference in sperm production, testicular morphology, and radioimmunoassay of serum testosterone levels compared to the control group. However, sperm motility was significantly reduced in rats under RAS inhibition. CONCLUSION: This finding was likely mediated by the identification of Ang receptors in the tails of rat sperm given that Ang receptors may play a role in the modulation of sperm motility. Identification of RAS-related proteins involved in sperm motility may help to explain their roles in motility. Our data provide general safety evidence for the male fertilization ability after paternal sacubitril/valsartan and ramipril exposure.
Collapse
|
23
|
DuPriest E, Hebert J, Morita M, Marek N, Meserve EEK, Andeen N, Houseman EA, Qi Y, Alwasel S, Nyengaard J, Morgan T. Fetal Renal DNA Methylation and Developmental Programming of Stress-Induced Hypertension in Growth-Restricted Male Mice. Reprod Sci 2020; 27:1110-1120. [PMID: 32046425 DOI: 10.1007/s43032-019-00121-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022]
Abstract
Fetal growth restriction (FGR) is associated with developmental programming of adult onset hypertension, which may be related to differences in nephron development. Prior studies showed that maternal nutrient restriction is associated with reduced nephrogenesis in rodents, especially in male progeny. We hypothesized that maternal genetic risk for FGR may similarly affect fetal kidney development, leading to adult onset hypertension. We employed an angiotensinogen (AGT) gene titration transgenic (TG) construct with 3 copies of the mouse AGT gene that mimics a common human genotype (AGT A[-6]G) associated with FGR. We investigated whether FGR in 2-copy (wild type, [WT]) progeny from 3-copy TG dams leads to developmental programming differences in kidney development and adult blood pressure compared with age- and sex-matched controls. Progeny were tested in the late fetal period (e17.5), neonatal period (2 weeks of age), and as young adults (12 weeks). We measured weights, tested for renal oxidative stress, compared renal DNA methylation profiles, counted the number of glomeruli, and measured adult blood pressure ± stress. Progeny from TG dams were growth restricted with evidence of renal oxidative stress, males showed fetal renal DNA hypermethylation, they had fewer glomeruli, and they developed stress-induced hypertension as adults. Their female siblings did not share this pathology and instead resembled progeny from WT dams. Surprisingly, glomerular counts in the neonatal period were not different between sexes or maternal genotypes. In turn, we suspect that differences in fetal renal DNA methylation may affect the long-term viability of glomeruli, rather than reducing nephrogenesis.
Collapse
Affiliation(s)
- Elizabeth DuPriest
- Departments of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.,Division of Natural Science and Health, Warner Pacific University, Portland, OR, USA
| | - Jessica Hebert
- Departments of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.,Department of Biology, Portland State University, Portland, OR, USA
| | - Mayu Morita
- Departments of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Nicole Marek
- Departments of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Emily E K Meserve
- Departments of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.,Department of Anatomic & Clinical Pathology, Maine Medical Center, Portland, ME, USA
| | - Nicole Andeen
- Departments of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - E Andres Houseman
- College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| | - Yue Qi
- Departments of Cardiovascular Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Saleh Alwasel
- Department of Zoology, King Saud University, Riyadh, Saudi Arabia
| | - Jens Nyengaard
- Core Centre for Molecular Morphology, Department of Clinical Medicine, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Terry Morgan
- Departments of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| |
Collapse
|
24
|
Steglich A, Hickmann L, Linkermann A, Bornstein S, Hugo C, Todorov VT. Beyond the Paradigm: Novel Functions of Renin-Producing Cells. Rev Physiol Biochem Pharmacol 2020; 177:53-81. [PMID: 32691160 DOI: 10.1007/112_2020_27] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The juxtaglomerular renin-producing cells (RPC) of the kidney are referred to as the major source of circulating renin. Renin is the limiting factor in renin-angiotensin system (RAS), which represents a proteolytic cascade in blood plasma that plays a central role in the regulation of blood pressure. Further cells disseminated in the entire organism express renin at a low level as part of tissue RASs, which are thought to locally modulate the effects of systemic RAS. In recent years, it became increasingly clear that the renal RPC are involved in developmental, physiological, and pathophysiological processes outside RAS. Based on recent experimental evidence, a novel concept emerges postulating that next to their traditional role, the RPC have non-canonical RAS-independent progenitor and renoprotective functions. Moreover, the RPC are part of a widespread renin lineage population, which may act as a global stem cell pool coordinating homeostatic, stress, and regenerative responses throughout the organism. This review focuses on the RAS-unrelated functions of RPC - a dynamic research area that increasingly attracts attention.
Collapse
Affiliation(s)
- Anne Steglich
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Linda Hickmann
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Andreas Linkermann
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stefan Bornstein
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Christian Hugo
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Vladimir T Todorov
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.
| |
Collapse
|
25
|
Tao XR, Rong JB, Lu HS, Daugherty A, Shi P, Ke CL, Zhang ZC, Xu YC, Wang JA. Angiotensinogen in hepatocytes contributes to Western diet-induced liver steatosis. J Lipid Res 2019; 60:1983-1995. [PMID: 31604805 PMCID: PMC6889717 DOI: 10.1194/jlr.m093252] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is considered as a liver manifestation of metabolic disorders. Previous studies indicate that the renin-angiotensin system (RAS) plays a complex role in NAFLD. As the only precursor of the RAS, decreased angiotensinogen (AGT) profoundly impacts RAS bioactivity. Here, we investigated the role of hepatocyte-derived AGT in liver steatosis. AGT floxed mice (hepAGT+/+) and hepatocyte-specific AGT-deficient mice (hepAGT−/−) were fed a Western diet and a normal laboratory diet for 12 weeks, respectively. Compared with hepAGT+/+ mice, Western diet-fed hepAGT−/− mice gained less body weight with improved insulin sensitivity. The attenuated severity of liver steatosis in hepAGT−/− mice was evidenced by histologic changes and reduced intrahepatic triglycerides. The abundance of SREBP1 and its downstream molecules, acetyl-CoA carboxylase and FASN, was suppressed in hepAGT−/− mice. Furthermore, serum derived from hepAGT+/+ mice stimulated hepatocyte SREBP1 expression, which could be diminished by protein kinase B (Akt)/mammalian target of rapamycin (mTOR) inhibition in vitro. Administration of losartan did not affect diet-induced body weight gain, liver steatosis severity, and hepatic p-Akt, p-mTOR, and SREBP1 protein abundance in hepAGT+/+ mice. These data suggest that attenuation of Western diet-induced liver steatosis in hepAGT−/− mice is associated with the alternation of the Akt/mTOR/SREBP-1c pathway.
Collapse
Affiliation(s)
- Xin-Ran Tao
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia-Bing Rong
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong S Lu
- Saha Cardiovascular Research Center and Departments of Pharmacology and Nutritional Sciences and Physiology, University of Kentucky, Lexington, KY
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Departments of Pharmacology and Nutritional Sciences and Physiology, University of Kentucky, Lexington, KY
| | - Peng Shi
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chang-Le Ke
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhao-Cai Zhang
- Department of Intensive Care Unit, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yin-Chuan Xu
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian-An Wang
- Department of Cardiology, Cardiovascular Key Laboratory of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
26
|
Abstract
Autosomal dominant tubulointerstitial kidney disease (ADTKD) is a recently defined entity that includes rare kidney diseases characterized by tubular damage and interstitial fibrosis in the absence of glomerular lesions, with inescapable progression to end-stage renal disease. These diseases have long been neglected and under-recognized, in part due to confusing and inconsistent terminology. The introduction of a gene-based, unifying terminology led to the identification of an increasing number of cases, with recent data suggesting that ADTKD is one of the more common monogenic kidney diseases after autosomal dominant polycystic kidney disease, accounting for ~5% of monogenic disorders causing chronic kidney disease. ADTKD is caused by mutations in at least five different genes, including UMOD, MUC1, REN, HNF1B and, more rarely, SEC61A1. These genes encode various proteins with renal and extra-renal functions. The mundane clinical characteristics and lack of appreciation of family history often result in a failure to diagnose ADTKD. This Primer highlights the different types of ADTKD and discusses the distinct genetic and clinical features as well as the underlying mechanisms.
Collapse
|
27
|
Affiliation(s)
- Victor J. Dzau
- From the Office of the President, National Academy of Medicine (formerly the Institute of Medicine), Washington, DC
| | - Celynne A. Balatbat
- From the Office of the President, National Academy of Medicine (formerly the Institute of Medicine), Washington, DC
| |
Collapse
|
28
|
Mkaouar H, Akermi N, Kriaa A, Abraham AL, Jablaoui A, Soussou S, Mokdad-Gargouri R, Maguin E, Rhimi M. Serine protease inhibitors and human wellbeing interplay: new insights for old friends. PeerJ 2019; 7:e7224. [PMID: 31531264 PMCID: PMC6718151 DOI: 10.7717/peerj.7224] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 05/31/2019] [Indexed: 12/14/2022] Open
Abstract
Serine Protease Inhibitors (Serpins) control tightly regulated physiological processes and their dysfunction is associated to various diseases. Thus, increasing interest is given to these proteins as new therapeutic targets. Several studies provided functional and structural data about human serpins. By comparison, only little knowledge regarding bacterial serpins exists. Through the emergence of metagenomic studies, many bacterial serpins were identified from numerous ecological niches including the human gut microbiota. The origin, distribution and function of these proteins remain to be established. In this report, we shed light on the key role of human and bacterial serpins in health and disease. Moreover, we analyze their function, phylogeny and ecological distribution. This review highlights the potential use of bacterial serpins to set out new therapeutic approaches.
Collapse
Affiliation(s)
- Héla Mkaouar
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Nizar Akermi
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Aicha Kriaa
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | | | - Amin Jablaoui
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Souha Soussou
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Raja Mokdad-Gargouri
- Laboratory of Molecular Biology of Eukaryotes, Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Emmanuelle Maguin
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Moez Rhimi
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| |
Collapse
|
29
|
Pandey KN. Genetic Ablation and Guanylyl Cyclase/Natriuretic Peptide Receptor-A: Impact on the Pathophysiology of Cardiovascular Dysfunction. Int J Mol Sci 2019; 20:ijms20163946. [PMID: 31416126 PMCID: PMC6721781 DOI: 10.3390/ijms20163946] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/09/2019] [Accepted: 08/10/2019] [Indexed: 12/11/2022] Open
Abstract
Mice bearing targeted gene mutations that affect the functions of natriuretic peptides (NPs) and natriuretic peptide receptors (NPRs) have contributed important information on the pathogenesis of hypertension, kidney disease, and cardiovascular dysfunction. Studies of mice having both complete gene disruption and tissue-specific gene ablation have contributed to our understanding of hypertension and cardiovascular disorders. These phenomena are consistent with an oligogenic inheritance in which interactions among a few alleles may account for genetic susceptibility to hypertension, renal insufficiency, and congestive heart failure. In addition to gene knockouts conferring increased risks of hypertension, kidney disorders, and cardiovascular dysfunction, studies of gene duplications have identified mutations that protect against high blood pressure and cardiovascular events, thus generating the notion that certain alleles can confer resistance to hypertension and heart disease. This review focuses on the intriguing phenotypes of Npr1 gene disruption and gene duplication in mice, with emphasis on hypertension and cardiovascular events using mouse models carrying Npr1 gene knockout and/or gene duplication. It also describes how Npr1 gene targeting in mice has contributed to our knowledge of the roles of NPs and NPRs in dose-dependently regulating hypertension and cardiovascular events.
Collapse
Affiliation(s)
- Kailash N Pandey
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
30
|
van Roeyen CRC, Martin IV, Drescher A, Schuett KA, Hermert D, Raffetseder U, Otten S, Buhl EM, Braun GS, Kuppe C, Liehn E, Boor P, Weiskirchen R, Eriksson U, Gross O, Eitner F, Floege J, Ostendorf T. Identification of platelet-derived growth factor C as a mediator of both renal fibrosis and hypertension. Kidney Int 2019; 95:1103-1119. [PMID: 30827511 DOI: 10.1016/j.kint.2018.11.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 11/15/2018] [Accepted: 11/21/2018] [Indexed: 02/06/2023]
Abstract
Platelet-derived growth factors (PDGF) have been implicated in kidney disease progression. We previously found that PDGF-C is upregulated at sites of renal fibrosis and that antagonism of PDGF-C reduces fibrosis in the unilateral ureteral obstruction model. We studied the role of PDGF-C in collagen 4A3-/- ("Alport") mice, a model of progressive renal fibrosis with greater relevance to human kidney disease. Alport mice were crossbred with PDGF-C-/- mice or administered a neutralizing PDGF-C antibody. Both PDGF-C deficiency and neutralization reduced serum creatinine and blood urea nitrogen levels and mitigated glomerular injury, renal fibrosis, and renal inflammation. Unexpectedly, systolic blood pressure was also reduced in both Alport and wild-type mice treated with a neutralizing PDGF-C antibody. Neutralization of PDGF-C reduced arterial wall thickness in the renal cortex of Alport mice. Aortic rings isolated from anti-PDGF-C-treated wildtype mice exhibited reduced tension and faster relaxation than those of untreated mice. In vitro, PDGF-C upregulated angiotensinogen in aortic tissue and in primary hepatocytes and induced nuclear factor κB (NFκB)/p65-binding to the angiotensinogen promoter in hepatocytes. Neutralization of PDGF-C suppressed transcript expression of angiotensinogen in Alport mice and angiotensin II receptor type 1 in Alport and wildtype mice. Finally, administration of neutralizing PDGF-C antibodies ameliorated angiotensin II-induced hypertension in healthy mice. Thus, in addition to its key role in mediating renal fibrosis, we identified PDGF-C as a mediator of hypertension via effects on renal vasculature and on the renin-angiotensin system. The contribution to both renal fibrosis and hypertension render PDGF-C an attractive target in progressive kidney disease.
Collapse
Affiliation(s)
- Claudia R C van Roeyen
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany.
| | - Ina V Martin
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Ana Drescher
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | | | - Daniela Hermert
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Ute Raffetseder
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Stephanie Otten
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Eva M Buhl
- Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Gerald S Braun
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Christoph Kuppe
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Elisa Liehn
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Peter Boor
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry RWTH Aachen University, Aachen, Germany
| | - Ulf Eriksson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Oliver Gross
- Division of Nephrology and Rheumatology, University Medicine Göttingen, Göttingen, Germany
| | - Frank Eitner
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Kidney Diseases Research, Bayer AG, Wuppertal, Germany
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Tammo Ostendorf
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| |
Collapse
|
31
|
Wu Y, Wang M, Zhang J, Sun N, Li C. A new model of the mechanism underlying lead poisoning: SNP in miRNA target region influence the AGT expression level. Hereditas 2019; 156:6. [PMID: 30700972 PMCID: PMC6348078 DOI: 10.1186/s41065-019-0084-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/20/2019] [Indexed: 11/25/2022] Open
Abstract
Background To determine if the rs7079 polymorphism located in the 3′ UTR of the angiotensinogen gene (AGT) altered AGT gene expression and the risk of lead poisoning. A case-control study and luciferase reporter gene assay identified a significant association between rs7079 variants and the risk of lead poisoning. Results Serum AGT levels were significantly higher in individuals carrying the rs7079 CA genotype, as compared to those carrying the rs7079 CC genotype. The binding of the miRNA mimics miR-31-5p and miR-584-5p to the 3′ UTR of AGT differed based on which rs7079 variant was present, implying that AGT gene expression depends on the rs7079 variant carried. Conclusions The rs7079 C to A substitution reduced the binding of miR-31-5p/miR-584-5p to the 3′ UTR of AGT, possibly altering the risk of lead poisoning.
Collapse
Affiliation(s)
- Yu Wu
- 1Wuxi Medical School, Jiangnan University, 499 Jincheng Rd, Wuxi, 214023 People's Republic of China
| | - Miaomiao Wang
- Department of Occupational Health, Wuxi Center for Disease Control and Prevention, Wuxi, China
| | - Jinlong Zhang
- Department of Occupational Health, Wuxi Center for Disease Control and Prevention, Wuxi, China
| | - Na Sun
- Department of Occupational Health, Wuxi Center for Disease Control and Prevention, Wuxi, China
| | - Chunping Li
- Department of Occupational Health, Wuxi Center for Disease Control and Prevention, Wuxi, China
| |
Collapse
|
32
|
Li X, Zhuang J, Uhal BD. Local activation of the pulmonary extravascular angiotensin system induces epithelial apoptosis and lung fibrosis. ACTA ACUST UNITED AC 2018; 5:192-200. [PMID: 32524006 DOI: 10.15406/jlprr.2018.05.00191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Previous work suggests that a local extravascular angiotensin system plays an important role in the development of pulmonary fibrosis through stimulation of alveolar epithelial cell (AEC) apoptosis and collagen deposition. To demonstrate a causative role for the local tissue angiotensin (ANG) system in lung fibrosis, we hypothesize that overexpression of the angiotensinogen (AGT) gene or pharmacologic elevation of lung tissue ANG II levels might cause apoptosis of AECs and lung fibrosis. ANGII levels were elevated in rat or mouse lung tissue by intratracheal instillation of either purified ANGII or an adenovirus expressing AGT, or by ubiquitous overexpression of AGT in transgenic mice. Intratracheal instillation of purified ANGII caused significant collagen accumulation in lung tissue, both ex vivo and in vivo. Ubiquitous overexpression of AGT enhanced the profibrotic effect of bleomycin given at suboptimal doses. Intratracheal delivery of an adenoviral vector expressing mouse AGT (Ad-AGT) overexpressed AGT primarily in AECs and caused both apoptosis of AECs and pulmonary fibrosis. The lung collagen accumulation and AEC apoptosis caused by Ad-AGT was blocked by the caspase inhibitor ZVAD-fmk, by the ANG receptor AT1 antagonist Losartan or by the non-selective ANGII receptor antagonist Saralasin. Together, these data support the hypothesis that elevated pulmonary expression of AGT and its conversion to angiotensin II plays a causative role in the development of lung fibrosis through its induction of AEC apoptosis.
Collapse
Affiliation(s)
- Xiaopeng Li
- Department of Pediatrics, Michigan State University, USA
| | - Jiaju Zhuang
- Department of Physiology, Bethune Military Medical College, China
| | - Bruce D Uhal
- Department of Pediatrics, Michigan State University, USA
| |
Collapse
|
33
|
Satou R, Penrose H, Navar LG. Inflammation as a Regulator of the Renin-Angiotensin System and Blood Pressure. Curr Hypertens Rep 2018; 20:100. [PMID: 30291560 DOI: 10.1007/s11906-018-0900-0] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW Mechanisms facilitating progression of hypertension via cross stimulation of the renin-angiotensin system (RAS) and inflammation have been proposed. Accordingly, we review and update evidence for regulation of RAS components by pro-inflammatory factors. RECENT FINDINGS Angiotensin II (Ang II), which is produced by RAS, induces vasoconstriction and consequent blood pressure elevation. In addition to this direct action, chronically elevated Ang II stimulates several pathophysiological mechanisms including generation of oxidative stress, stimulation of the nervous system, alterations in renal hemodynamics, and activation of the immune system. In particular, an activated immune system has been shown to contribute to the development of hypertension. Recent studies have demonstrated that immune cell-derived pro-inflammatory cytokines regulate RAS components, further accelerating systemic and local Ang II formation. Specifically, regulation of angiotensinogen (AGT) production by pro-inflammatory cytokines in the liver and kidney is proposed as a key mechanism underlying the progression of Ang II-dependent hypertension.
Collapse
Affiliation(s)
- Ryousuke Satou
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, 1430 Tulane Avenue, SL39, New Orleans, LA, 70112-2699, USA.
| | - Harrison Penrose
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, 1430 Tulane Avenue, SL39, New Orleans, LA, 70112-2699, USA
| | - L Gabriel Navar
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, 1430 Tulane Avenue, SL39, New Orleans, LA, 70112-2699, USA
| |
Collapse
|
34
|
Pandey KN. Molecular and genetic aspects of guanylyl cyclase natriuretic peptide receptor-A in regulation of blood pressure and renal function. Physiol Genomics 2018; 50:913-928. [PMID: 30169131 DOI: 10.1152/physiolgenomics.00083.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Natriuretic peptides (NPs) exert diverse effects on several biological and physiological systems, such as kidney function, neural and endocrine signaling, energy metabolism, and cardiovascular function, playing pivotal roles in the regulation of blood pressure (BP) and cardiac and vascular homeostasis. NPs are collectively known as anti-hypertensive hormones and their main functions are directed toward eliciting natriuretic/diuretic, vasorelaxant, anti-proliferative, anti-inflammatory, and anti-hypertrophic effects, thereby, regulating the fluid volume, BP, and renal and cardiovascular conditions. Interactions of NPs with their cognate receptors display a central role in all aspects of cellular, biochemical, and molecular mechanisms that govern physiology and pathophysiology of BP and cardiovascular events. Among the NPs atrial and brain natriuretic peptides (ANP and BNP) activate guanylyl cyclase/natriuretic peptide receptor-A (GC-A/NPRA) and initiate intracellular signaling. The genetic disruption of Npr1 (encoding GC-A/NPRA) in mice exhibits high BP and hypertensive heart disease that is seen in untreated hypertensive subjects, including high BP and heart failure. There has been a surge of interest in the NPs and their receptors and a wealth of information have emerged in the last four decades, including molecular structure, signaling mechanisms, altered phenotypic characterization of transgenic and gene-targeted animal models, and genetic analyses in humans. The major goal of the present review is to emphasize and summarize the critical findings and recent discoveries regarding the molecular and genetic regulation of NPs, physiological metabolic functions, and the signaling of receptor GC-A/NPRA with emphasis on the BP regulation and renal and cardiovascular disorders.
Collapse
Affiliation(s)
- Kailash N Pandey
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine , New Orleans, Louisiana
| |
Collapse
|
35
|
Intratubular and intracellular renin-angiotensin system in the kidney: a unifying perspective in blood pressure control. Clin Sci (Lond) 2018; 132:1383-1401. [PMID: 29986878 DOI: 10.1042/cs20180121] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/05/2018] [Accepted: 06/13/2018] [Indexed: 12/18/2022]
Abstract
The renin-angiotensin system (RAS) is widely recognized as one of the most important vasoactive hormonal systems in the physiological regulation of blood pressure and the development of hypertension. This recognition is derived from, and supported by, extensive molecular, cellular, genetic, and pharmacological studies on the circulating (tissue-to-tissue), paracrine (cell-to-cell), and intracrine (intracellular, mitochondrial, nuclear) RAS during last several decades. Now, it is widely accepted that circulating and local RAS may act independently or interactively, to regulate sympathetic activity, systemic and renal hemodynamics, body salt and fluid balance, and blood pressure homeostasis. However, there remains continuous debate with respect to the specific sources of intratubular and intracellular RAS in the kidney and other tissues, the relative contributions of the circulating RAS to intratubular and intracellular RAS, and the roles of intratubular compared with intracellular RAS to the normal control of blood pressure or the development of angiotensin II (ANG II)-dependent hypertension. Based on a lecture given at the recent XI International Symposium on Vasoactive Peptides held in Horizonte, Brazil, this article reviews recent studies using mouse models with global, kidney- or proximal tubule-specific overexpression (knockin) or deletion (knockout) of components of the RAS or its receptors. Although much knowledge has been gained from cell- and tissue-specific transgenic or knockout models, a unifying and integrative approach is now required to better understand how the circulating and local intratubular/intracellular RAS act independently, or with other vasoactive systems, to regulate blood pressure, cardiovascular and kidney function.
Collapse
|
36
|
Wu CH, Mohammadmoradi S, Chen JZ, Sawada H, Daugherty A, Lu HS. Renin-Angiotensin System and Cardiovascular Functions. Arterioscler Thromb Vasc Biol 2018; 38:e108-e116. [PMID: 29950386 PMCID: PMC6039412 DOI: 10.1161/atvbaha.118.311282] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Chia-Hua Wu
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., A.D., H.S.L.)
| | - Shayan Mohammadmoradi
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., A.D., H.S.L.)
| | - Jeff Z Chen
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Physiology (J.Z.C., A.D., H.S.L.), University of Kentucky, Lexington
| | - Hisashi Sawada
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., A.D., H.S.L.)
- Department of Physiology (J.Z.C., A.D., H.S.L.), University of Kentucky, Lexington
| | - Hong S Lu
- From the Saha Cardiovascular Research Center (C.-H.W., S.M., J.Z.C., H.S., A.D., H.S.L.)
- Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., A.D., H.S.L.)
- Department of Physiology (J.Z.C., A.D., H.S.L.), University of Kentucky, Lexington
| |
Collapse
|
37
|
Nishiyama A, Kobori H. Independent regulation of renin-angiotensin-aldosterone system in the kidney. Clin Exp Nephrol 2018; 22:1231-1239. [PMID: 29600408 PMCID: PMC6163102 DOI: 10.1007/s10157-018-1567-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 03/21/2018] [Indexed: 01/13/2023]
Abstract
Renin-angiotensin-aldosterone system (RAAS) plays important roles in regulating renal hemodynamics and functions, as well as in the pathophysiology of hypertension and renal disease. In the kidney, angiotensin II (Ang II) production is controlled by independent multiple mechanisms. Ang II is compartmentalized in the renal interstitial fluid with much higher concentrations than those existing in the circulation. Inappropriate activation of the intrarenal RAAS is an important contributor to the pathogenesis of hypertension and renal injury. It has been revealed that intrarenal Ang II levels are predominantly regulated by angiotensinogen and therefore, urinary angiotensinogen could be a biomarker for intrarenal Ang II generation. In addition, recent studies have demonstrated that aldosterone contributes to the progression of renal injury via direct actions on glomerular podocytes, mesangial cells, proximal tubular cells and tubulo-interstitial fibroblasts through the activation of locally expressed mineralocorticoid receptor. Thus, it now appears that intrarenal RAAS is independently regulated and its inappropriate activation contributes to the pathogenesis of the development of hypertension and renal disease. This short review article will focus on the independent regulation of the intrarenal RAAS with an emphasis on the specific role of angiotensinogen.
Collapse
Affiliation(s)
- Akira Nishiyama
- Department of Pharmacology, Faculty of Medicine, Kagawa University, 1750-1 Miki-cho, Kita-gun, Kagawa, 761-0793, Japan.
| | - Hiroyuki Kobori
- Departments of Pharmacology and Nephrology, Faculty of Medicine, International University of Health and Welfare, Narita, Japan
| |
Collapse
|
38
|
Abstract
Angiotensin-converting enzyme (ACE) - a zinc-dependent dicarboxypeptidase with two catalytic domains - plays a major part in blood pressure regulation by converting angiotensin I to angiotensin II. However, ACE cleaves many peptides besides angiotensin I and thereby affects diverse physiological functions, including renal development and male reproduction. In addition, ACE has a role in both innate and adaptive responses by modulating macrophage and neutrophil function - effects that are magnified when these cells overexpress ACE. Macrophages that overexpress ACE are more effective against tumours and infections. Neutrophils that overexpress ACE have an increased production of superoxide, which increases their ability to kill bacteria. These effects are due to increased ACE activity but are independent of angiotensin II. ACE also affects the display of major histocompatibility complex (MHC) class I and MHC class II peptides, potentially by enzymatically trimming these peptides. Understanding how ACE expression and activity affect myeloid cells may hold great promise for therapeutic manipulation, including the treatment of both infection and malignancy.
Collapse
|
39
|
Bernstein KE, Khan Z, Giani JF, Cao DY, Bernstein EA, Shen XZ. Angiotensin-converting enzyme in innate and adaptive immunity. Nat Rev Nephrol 2018; 14:325-336. [PMID: 29578208 DOI: 10.1038/nrneph.2018.15] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Angiotensin-converting enzyme (ACE) - a zinc-dependent dicarboxypeptidase with two catalytic domains - plays a major part in blood pressure regulation by converting angiotensin I to angiotensin II. However, ACE cleaves many peptides besides angiotensin I and thereby affects diverse physiological functions, including renal development and male reproduction. In addition, ACE has a role in both innate and adaptive responses by modulating macrophage and neutrophil function - effects that are magnified when these cells overexpress ACE. Macrophages that overexpress ACE are more effective against tumours and infections. Neutrophils that overexpress ACE have an increased production of superoxide, which increases their ability to kill bacteria. These effects are due to increased ACE activity but are independent of angiotensin II. ACE also affects the display of major histocompatibility complex (MHC) class I and MHC class II peptides, potentially by enzymatically trimming these peptides. Understanding how ACE expression and activity affect myeloid cells may hold great promise for therapeutic manipulation, including the treatment of both infection and malignancy.
Collapse
Affiliation(s)
- Kenneth E Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Zakir Khan
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
| | - Jorge F Giani
- Department of Biomedical Sciences, Cedars-Sinai Medical Center.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Duo-Yao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
| | | | - Xiao Z Shen
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
40
|
Ferrario CM, Mullick AE. Renin angiotensin aldosterone inhibition in the treatment of cardiovascular disease. Pharmacol Res 2017; 125:57-71. [PMID: 28571891 PMCID: PMC5648016 DOI: 10.1016/j.phrs.2017.05.020] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/11/2017] [Accepted: 05/15/2017] [Indexed: 02/07/2023]
Abstract
A collective century of discoveries establishes the importance of the renin angiotensin aldosterone system in maintaining blood pressure, fluid volume and electrolyte homeostasis via autocrine, paracrine and endocrine signaling. While research continues to yield new functions of angiotensin II and angiotensin-(1-7), the gap between basic research and clinical application of these new findings is widening. As data accumulates on the efficacy of angiotensin converting enzyme inhibitors and angiotensin II receptor blockers as drugs of fundamental importance in the treatment of cardiovascular and renal disorders, it is becoming apparent that the achieved clinical benefits is suboptimal and surprisingly no different than what can be achieved with other therapeutic interventions. We discuss this issue and summarize new pathways and mechanisms effecting the synthesis and actions of angiotensin II. The presence of renin-independent non-canonical pathways for angiotensin II production are largely unaffected by agents inhibiting renin angiotensin system activity. Hence, new efforts should be directed to develop drugs that can effectively block the synthesis and/or action of intracellular angiotensin II. Improved drug penetration into cardiac or renal sites of disease, inhibiting chymase the primary angiotensin II forming enzyme in the human heart, and/or inhibiting angiotensinogen synthesis would all be more effective strategies to inhibit the system. Additionally, given the role of angiotensin II in the maintenance of renal homeostatic mechanisms, any new inhibitor should possess greater selectivity of targeting pathogenic angiotensin II signaling processes and thereby limit inappropriate inhibition.
Collapse
Affiliation(s)
- Carlos M Ferrario
- Department of Surgery, Wake Forest University Health Science, Medical Center Blvd., Winston Salem, NC 27157, United States.
| | - Adam E Mullick
- Cardiovascular Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, United States
| |
Collapse
|
41
|
Li XC, Zhang J, Zhuo JL. The vasoprotective axes of the renin-angiotensin system: Physiological relevance and therapeutic implications in cardiovascular, hypertensive and kidney diseases. Pharmacol Res 2017; 125:21-38. [PMID: 28619367 DOI: 10.1016/j.phrs.2017.06.005] [Citation(s) in RCA: 281] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/08/2017] [Accepted: 06/09/2017] [Indexed: 01/11/2023]
Abstract
The renin-angiotensin system (RAS) is undisputedly one of the most prominent endocrine (tissue-to-tissue), paracrine (cell-to-cell) and intracrine (intracellular/nuclear) vasoactive systems in the physiological regulation of neural, cardiovascular, blood pressure, and kidney function. The importance of the RAS in the development and pathogenesis of cardiovascular, hypertensive and kidney diseases has now been firmly established in clinical trials and practice using renin inhibitors, angiotensin-converting enzyme (ACE) inhibitors, type 1 (AT1) angiotensin II (ANG II) receptor blockers (ARBs), or aldosterone receptor antagonists as major therapeutic drugs. The major mechanisms of actions for these RAS inhibitors or receptor blockers are mediated primarily by blocking the detrimental effects of the classic angiotensinogen/renin/ACE/ANG II/AT1/aldosterone axis. However, the RAS has expanded from this classic axis to include several other complex biochemical and physiological axes, which are derived from the metabolism of this classic axis. Currently, at least five axes of the RAS have been described, with each having its key substrate, enzyme, effector peptide, receptor, and/or downstream signaling pathways. These include the classic angiotensinogen/renin/ACE/ANG II/AT1 receptor, the ANG II/APA/ANG III/AT2/NO/cGMP, the ANG I/ANG II/ACE2/ANG (1-7)/Mas receptor, the prorenin/renin/prorenin receptor (PRR or Atp6ap2)/MAP kinases ERK1/2/V-ATPase, and the ANG III/APN/ANG IV/IRAP/AT4 receptor axes. Since the roles and therapeutic implications of the classic angiotensinogen/renin/ACE/ANG II/AT1 receptor axis have been extensively reviewed, this article will focus primarily on reviewing the roles and therapeutic implications of the vasoprotective axes of the RAS in cardiovascular, hypertensive and kidney diseases.
Collapse
Affiliation(s)
- Xiao C Li
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology, Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216-4505, USA
| | - Jianfeng Zhang
- Department of Emergency Medicine, The 2nd Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Jia L Zhuo
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology, Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216-4505, USA.
| |
Collapse
|
42
|
Chmelař J, Kotál J, Langhansová H, Kotsyfakis M. Protease Inhibitors in Tick Saliva: The Role of Serpins and Cystatins in Tick-host-Pathogen Interaction. Front Cell Infect Microbiol 2017; 7:216. [PMID: 28611951 PMCID: PMC5447049 DOI: 10.3389/fcimb.2017.00216] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 05/11/2017] [Indexed: 11/23/2022] Open
Abstract
The publication of the first tick sialome (salivary gland transcriptome) heralded a new era of research of tick protease inhibitors, which represent important constituents of the proteins secreted via tick saliva into the host. Three major groups of protease inhibitors are secreted into saliva: Kunitz inhibitors, serpins, and cystatins. Kunitz inhibitors are anti-hemostatic agents and tens of proteins with one or more Kunitz domains are known to block host coagulation and/or platelet aggregation. Serpins and cystatins are also anti-hemostatic effectors, but intriguingly, from the translational perspective, also act as pluripotent modulators of the host immune system. Here we focus especially on this latter aspect of protease inhibition by ticks and describe the current knowledge and data on secreted salivary serpins and cystatins and their role in tick-host-pathogen interaction triad. We also discuss the potential therapeutic use of tick protease inhibitors.
Collapse
Affiliation(s)
- Jindřich Chmelař
- Faculty of Science, University of South Bohemia in České BudějoviceČeské Budějovice, Czechia
| | - Jan Kotál
- Faculty of Science, University of South Bohemia in České BudějoviceČeské Budějovice, Czechia.,Institute of Parasitology, Biology Center, Czech Academy of SciencesČeské Budějovice, Czechia
| | - Helena Langhansová
- Faculty of Science, University of South Bohemia in České BudějoviceČeské Budějovice, Czechia.,Institute of Parasitology, Biology Center, Czech Academy of SciencesČeské Budějovice, Czechia
| | - Michail Kotsyfakis
- Institute of Parasitology, Biology Center, Czech Academy of SciencesČeské Budějovice, Czechia
| |
Collapse
|
43
|
Oka M, Medrano S, Sequeira-Lόpez MLS, Gómez RA. Chronic Stimulation of Renin Cells Leads to Vascular Pathology. Hypertension 2017; 70:119-128. [PMID: 28533331 DOI: 10.1161/hypertensionaha.117.09283] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/09/2017] [Accepted: 04/04/2017] [Indexed: 01/14/2023]
Abstract
Experimental or spontaneous genomic mutations of the renin-angiotensin system or its pharmacological inhibition in early life leads to renal abnormalities, including poorly developed renal medulla, papillary atrophy, hydronephrosis, inability to concentrate the urine, polyuria, polydipsia, renal failure, and anemia. At the core of such complex phenotype is the presence of unique vascular abnormalities: the renal arterioles do not branch or elongate properly and they have disorganized, concentric hypertrophy. This lesion has been puzzling because it is often found in hypertensive individuals whereas mutant or pharmacologically inhibited animals are hypotensive. Remarkably, when renin cells are ablated with diphtheria toxin, the vascular hypertrophy does not occur, suggesting that renin cells per se may contribute to the vascular disease. To test this hypothesis, on a Ren1c-/- background, we generated mutant mice with reporter expression (Ren1c-/-;Ren1c-Cre;R26R.mTmG and Ren1c-/-;Ren1c-Cre;R26R.LacZ) to trace the fate of reninnull cells. To assess whether reninnull cells maintain their renin promoter active, we used Ren1c-/-;Ren1c-YFP mice that transcribe YFP (yellow fluorescent protein) directed by the renin promoter. We also followed the expression of Akr1b7 and miR-330-5p, markers of cells programmed for the renin phenotype. Contrary to what we expected, reninnull cells did not die or disappear. Instead, they survived, increased in number along the renal arterial tree, and maintained an active molecular memory of the myoepitheliod renin phenotype. Furthermore, null cells of the renin lineage occupied the walls of the arteries and arterioles in a chaotic, directionless pattern directly contributing to the concentric arterial hypertrophy.
Collapse
Affiliation(s)
- Masafumi Oka
- From the Department of Pediatrics, University of Virginia, Charlottesville
| | - Silvia Medrano
- From the Department of Pediatrics, University of Virginia, Charlottesville
| | | | - R Ariel Gómez
- From the Department of Pediatrics, University of Virginia, Charlottesville.
| |
Collapse
|
44
|
Rana A, Jain S, Puri N, Kaw M, Sirianni N, Eren D, Mopidevi BR, Kumar A. The transcriptional regulation of the human angiotensinogen gene after high-fat diet is haplotype-dependent: Novel insights into the gene-regulatory networks and implications for human hypertension. PLoS One 2017; 12:e0176373. [PMID: 28467442 PMCID: PMC5415177 DOI: 10.1371/journal.pone.0176373] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 04/09/2017] [Indexed: 11/18/2022] Open
Abstract
Single nucleotide polymorphisms (SNPs) in the human angiotensinogen (hAGT) gene may modulate its transcription and affect the regulation of blood pressure via activation of the renin-angiotensin aldosterone system (RAAS). In this regard, we have identified polymorphisms in the 2.5 Kb promoter of the hAGT gene that form two haplotype (Hap) blocks: -6A/G (-1670A/G, -1562C/T, -1561T/C) and -217A/G (-532T/C, -793A/G, -1074T/C & -1178G/A). hAGT gene with Hap -6A/-217A (Hap I) is associated with increased blood pressure whereas, Hap -6G/-217G (Hap II) is associated with normal blood pressure in human subjects. Since RAAS over activity contributes to hypertension in obesity, we have made transgenic mice (TG) containing either Hap I or Hap II of the hAGT gene to understand the role of obesity on its transcriptional regulation. Although, a high-fat diet (60% Kcal from fat, 12 weeks) elevates hAGT and mAGT regardless of haplotype, this effect is significantly (p<0.05) accentuated in Hap I mice, in both adipose and liver tissues. Chromatin Immuno- precipitation (ChIP) assay shows an increased binding of transcription factors including, GR, CEBPβ and STAT3 to the chromatin of the Hap I TG mice after high-fat diet as compared to Hap II TG mice (p<0.05). Differential plasma levels of hAGT in Hap II and I mice, after high-fat diet, further corroborate the variable transcriptional regulation of the hAGT, governed by gene-haplotypes. Taken together, our results show that SNPs in the Hap-I of the hAGT gene promote high-fat diet-induced binding of transcription factors GR, CEBP-β and STAT3, which lead to elevated expression of the hAGT gene in hepatic and adipose tissues.
Collapse
Affiliation(s)
- Anita Rana
- Department of Physiology and Pharmacology, University Of Toledo Medical Centre, Toledo, Ohio, United States of America
| | - Sudhir Jain
- Department of Physiology and Pharmacology, University Of Toledo Medical Centre, Toledo, Ohio, United States of America
| | - Nitin Puri
- Department of Physiology and Pharmacology, University Of Toledo Medical Centre, Toledo, Ohio, United States of America
| | - Meenakshi Kaw
- Department of Physiology and Pharmacology, University Of Toledo Medical Centre, Toledo, Ohio, United States of America
| | - Natalie Sirianni
- Department of Physiology and Pharmacology, University Of Toledo Medical Centre, Toledo, Ohio, United States of America
| | - Deniz Eren
- Department of Physiology and Pharmacology, University Of Toledo Medical Centre, Toledo, Ohio, United States of America
| | - Brahma Raju Mopidevi
- Department of Physiology and Pharmacology, University Of Toledo Medical Centre, Toledo, Ohio, United States of America
| | - Ashok Kumar
- Department of Physiology and Pharmacology, University Of Toledo Medical Centre, Toledo, Ohio, United States of America
- Department of Pathology, New York Medical College, Valhalla, New York, United States of America
- * E-mail:
| |
Collapse
|
45
|
Kashihara T, Nakada T, Kojima K, Takeshita T, Yamada M. Angiotensin II activates Ca V 1.2 Ca 2+ channels through β-arrestin2 and casein kinase 2 in mouse immature cardiomyocytes. J Physiol 2017; 595:4207-4225. [PMID: 28295363 DOI: 10.1113/jp273883] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/10/2017] [Indexed: 12/25/2022] Open
Abstract
KEY POINTS Angiotensin II (AngII) is crucial in cardiovascular regulation in perinatal mammalians. Here we show that AngII increases twitch Ca2+ transients of mouse immature but not mature cardiomyocytes by robustly activating CaV 1.2 L-type Ca2+ channels through a novel signalling pathway involving angiotensin type 1 (AT1 ) receptors, β-arrestin2 and casein kinase 2. A β-arrestin-biased AT1 receptor agonist, TRV027, was as effective as AngII in activating L-type Ca2+ channels. Our results help understand the molecular mechanism by which AngII regulates the perinatal circulation and also suggest that β-arrestin-biased AT1 receptor agonists may be valuable therapeutics for paediatric heart failure. ABSTRACT Angiotensin II (AngII), the main effector peptide of the renin-angiotensin system, plays important roles in cardiovascular regulation in the perinatal period. Despite the well-known stimulatory effect of AngII on vascular contraction, little is known about regulation of contraction of the immature heart by AngII. Here we found that AngII significantly increased the peak amplitude of twitch Ca2+ transients by robustly activating L-type CaV 1.2 Ca2+ (CaV 1.2) channels in mouse immature but not mature cardiomyocytes. This response to AngII was mediated by AT1 receptors and β-arrestin2. A β-arrestin-biased AT1 receptor agonist was as effective as AngII in activating CaV 1.2 channels. Src-family tyrosine kinases (SFKs) and casein kinase 2α'β (CK2α'β) were sequentially activated when AngII activated CaV 1.2 channels. A cyclin-dependent kinase inhibitor, p27Kip1 (p27), inhibited CK2α'β, and AngII removed this inhibitory effect through phosphorylating tyrosine 88 of p27 via SFKs in cardiomyocytes. In a human embryonic kidney cell line, tsA201 cells, overexpression of CK2α'β but not c-Src directly activated recombinant CaV 1.2 channels composed of C-terminally truncated α1C , the distal C-terminus of α1C , β2C and α2 δ1 subunits, by phosphorylating threonine 1704 located at the interface between the proximal and the distal C-terminus of CaV 1.2α1C subunits. Co-immunoprecipitation revealed that CaV 1.2 channels, CK2α'β and p27 formed a macromolecular complex. Therefore, stimulation of AT1 receptors by AngII activates CaV 1.2 channels through β-arrestin2 and CK2α'β, thereby probably exerting a positive inotropic effect in the immature heart. Our results also indicated that β-arrestin-biased AT1 receptor agonists may be used as valuable therapeutics for paediatric heart failure in the future.
Collapse
Affiliation(s)
- Toshihide Kashihara
- Department of Molecular Pharmacology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Tsutomu Nakada
- Department of Molecular Pharmacology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Katsuhiko Kojima
- Department of Microbiology and Immunology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Toshikazu Takeshita
- Department of Microbiology and Immunology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Mitsuhiko Yamada
- Department of Molecular Pharmacology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| |
Collapse
|
46
|
Epochs in the depressor/pressor balance of the renin-angiotensin system. Clin Sci (Lond) 2017; 130:761-71. [PMID: 27128801 DOI: 10.1042/cs20150939] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/01/2016] [Indexed: 12/17/2022]
Abstract
The renin-angiotensin system (RAS) plays a commanding role in the regulation of extracellular fluid homoeostasis. Tigerstadt and Bergman first identified the RAS more than two centuries ago. By the 1980s a voyage of research and discovery into the mechanisms and actions of this system led to the development of drugs that block the RAS, which have become the mainstay for the treatment of cardiovascular and renal disease. In the last 25 years new components of the RAS have come to light, including the angiotensin type 2 receptor (AT2R) and the angiotensin-converting enzyme 2 (ACE2)/angiotensin-(1-7) [Ang(1-7)]/Mas receptor (MasR) axis. These have been shown to counter the classical actions of angiotensin II (AngII) at the predominant angiotensin type 1 receptor (AT1R). Our studies, and those of others, have demonstrated that targeting these depressor RAS pathways may be therapeutically beneficial. It is apparent that the evolution of both the pressor and depressor RAS pathways is distinct throughout life and that the depressor/pressor balance of the RAS vary between the sexes. These temporal patterns of expression suggest that therapies targeting the RAS could be optimized for discrete epochs in life.
Collapse
|
47
|
Coffman TM, Audoly LP, Oliverio MI. Review: Gene targeting studies of angiotensin II type 1 (AT1) receptors. J Renin Angiotensin Aldosterone Syst 2017; 2:S10-S15. [PMID: 28095241 DOI: 10.1177/14703203010020010201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Thomas M Coffman
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham NC, USA,
| | - Laurent P Audoly
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham NC, USA
| | - Michael I Oliverio
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham NC, USA
| |
Collapse
|
48
|
Gomez RA, Sequeira-Lopez MLS. Novel Functions of Renin Precursors in Homeostasis and Disease. Physiology (Bethesda) 2017; 31:25-33. [PMID: 26661526 DOI: 10.1152/physiol.00039.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Renin progenitors appear early and are found in multiple tissues throughout the embryo. Besides their well known role in blood pressure and fluid homeostasis, renin progenitors participate in tissue morphogenesis, repair, and regeneration, and may integrate immune and endocrine responses. In the bone marrow, renin cells offer clues to understand normal and neoplastic hematopoiesis.
Collapse
Affiliation(s)
- R Ariel Gomez
- University of Virginia School of Medicine, Child Health Research Center, Charlottesville, Virginia
| | | |
Collapse
|
49
|
Lin CH, Liao CC, Huang CH, Tung YT, Chang HC, Hsu MC, Huang CC. Proteomics Analysis to Identify and Characterize the Biomarkers and Physical Activities of Non-Frail and Frail Older Adults. Int J Med Sci 2017; 14:231-239. [PMID: 28367083 PMCID: PMC5370285 DOI: 10.7150/ijms.17627] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 12/28/2016] [Indexed: 11/05/2022] Open
Abstract
Globally, the proportion of older adults is increasing. Older people face chronic conditions such as sarcopenia and functional decline, which are often associated with disability and frailty. Proteomics assay of potential serum biomarkers of frailty in older adults. Older adults were divided into non-frail and frail groups (n = 6 each; 3 males in each group) in accordance with the Chinese-Canadian Study of Health and Aging Clinical Frailty Scale. Adults were measured for grip power and the 6-min walk test for physical activity, and venous blood was sampled after adults fasted for 8 h. Ultra-high-performance liquid chromatography-tandem mass spectrometry was used for proteomics assay. The groups were compared for levels of biomarkers by t test and Pearson correlation analysis. Non-frail and frail subjects had mean age 77.5±0.4 and 77.7±1.6 years, mean height 160.5±1.3 and 156.6±2.9 cm and mean weight 62.5±1.2 and 62.8±2.9 kg, respectively. Physical activity level was lower for frail than non-frail subjects (grip power: 13.8±0.4 vs 26.1±1.2 kg; 6-min walk test: 215.2±17.2 vs 438.3±17.2 m). Among 226 proteins detected, for 31, serum levels were significantly higher for frail than non-frail subjects; serum levels of Ig kappa chain V-III region WOL, COX7A2, and albumin were lower. The serum levels of ANGT, KG and AT were 2.05-, 1.76- and 2.22-fold lower (all p < 0.05; Figure 1A, 2A and 3A) for non-frail than frail subjects and were highly correlated with grip power (Figure 1B, 2B and 3B). Our study found that ANGT, KG and AT levels are known to increase with aging, so degenerated vascular function might be associated with frailty. In total, 226 proteins were revealed proteomics assay; levels of angiotensinogen (ANGT), kininogen-1 (KG) and antithrombin III (AT) were higher in frail than non-frail subjects (11.26±2.21 vs 5.09±0.74; 18.42±1.36 vs 11.64±1.36; 22.23±1.64 vs 9.52±0.95, respectively, p < 0.05). These 3 factors were highly correlated with grip power (p < 0.05), with higher correlations between grip power and serum levels of ANGT (r = -0.89), KG (r = -0.90), and AT (r = -0.84). In conclusion, this is the first study to demonstrate a serum proteomic profile characteristic of frailty in older adults. Serum ANGT, KG and AT levels could be potential biomarkers for monitoring the development and progression of frailty in older adults.
Collapse
Affiliation(s)
- Ching-Hung Lin
- Physical Education Office, Yuan Ze University, Taoyuan 32003, Taiwan
| | - Chen-Chung Liao
- Proteomics Research Center, National Yang-Ming University, Taipei 11221, Taiwan
| | - Chi-Huang Huang
- Department of Athletic Training and Health, National Taiwan Sport University, Taoyuan 33301, Taiwan
| | - Yu-Tang Tung
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan 33301, Taiwan
| | - Huan-Cheng Chang
- Department of Family Medicine, Taiwan Landseed Hospital, Ping-Jen City, Taoyuan 32449, Taiwan
| | - Mei-Chich Hsu
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan 33301, Taiwan;; Department of Sports Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;; Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chi-Chang Huang
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan 33301, Taiwan
| |
Collapse
|
50
|
Gnanasambandan R, Elumalai R, Soundararajan P, Lakkakula BVKS. Angiotensinogen gene polymorphisms and progression of chronic kidney disease in ADPKD patients. Clin Exp Nephrol 2016; 20:561-568. [PMID: 26482465 DOI: 10.1007/s10157-015-1183-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/07/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is a multisystemic and progressive disorder characterized by cyst formation and kidney enlargement and ultimately renal failure. Reduction of CKD progression in the ADPKD by pharmacological blockade of the renin-angiotensin-aldosterone system (RAAS) using ACE inhibitors indicated the involvement of RAAS pathway in the progression of CKD. The aim of the present study is to investigate the role of angiotensinogen tag-single nucleotide polymorphisms (AGT tag-SNPs) in progression of CKD. METHODS Twelve AGT tag-SNPs were genotyped in 102 ADPKD patients and 106 non-ADPKD subjects using FRET-based KASPar method. Genotypes and haplotypes were compared between ADPKD and controls. The effect of genotypes and hypertension on CKD progression was assessed using univariate and multivariate logistic regression. Mantel-Haenszel (M-H) stratified analysis was performed to study the interaction between CKD stages and hypertension. RESULTS Of the twelve tag-SNPs analyzed, only rs11122578 SNP deviated Hardy-Weinberg equilibrium in controls. Significant association between two AGT polymorphisms (rs11122577 and rs4762) and ADPKD was observed. Analysis of linkage disequilibrium revealed two haplotype blocks and haplotypes are not associated with ADPKD. The univariate analysis revealed that the age, hypertension, family history of diabetes and AGT rs4762 contributed to the progression of CKD in ADPKD. The modifier effect of these factors remained even after controlling other variables in multivariate analysis. CONCLUSIONS The results of our study suggest significant association between Thr207Met polymorphism of AGT and CKD progression and acts as an effect modifier of renal disease progression in ADPKD.
Collapse
Affiliation(s)
| | | | | | - Bhaskar V K S Lakkakula
- Department of Biomedical Sciences, Sri Ramachandra University, Chennai, India.
- Genetic Lab, Department of Biochemistry, Sickle Cell Institute Chhattisgarh, Pt. JNM Medical College, Raipur, C.G., India.
| |
Collapse
|