1
|
Rodrigues AF, Todiras M, Qadri F, Alenina N, Bader M. Angiotensin deficient FVB/N mice are normotensive. Br J Pharmacol 2023; 180:1843-1861. [PMID: 36740662 DOI: 10.1111/bph.16051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/17/2023] [Accepted: 01/31/2023] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND PURPOSE All previous rodent models lacking the peptide hormone angiotensin II (Ang II) were hypotensive. A mixed background strain with global deletion of the angiotensinogen gene was backcrossed to the FVB/N background (Agt-KO), a strain preferred for transgenic generation. Surprisingly, the resulting line turned out to be normotensive. Therefore, this study aimed to understand the unique blood pressure regulation of FVB/N mice without angiotensin peptides. EXPERIMENTAL APPROACH Acute and chronic recordings of blood pressure (BP) in freely-moving adult mice were performed to establish baseline BP. The pressure responses to sympatholytic and sympathomimetic as well as a nitric oxide inhibitor and donor compounds were used to quantify the neurogenic tone and endothelial function. The role of the renal nerves on baseline BP maintenance was tested by renal denervation. Finally, further phenotyping was done by gene expression analysis, histology and measurement of metabolites in plasma, urine and tissues. KEY RESULTS Baseline BP in adult FVB/N Agt-KO was unexpectedly unaltered. As compensatory mechanisms Agt-KO presented an increased sympathetic nerve activity and reduced endothelial nitric oxide production. However, FVB/N Agt-KO exhibited the renal morphological and physiological alterations previously found in mice lacking the production of Ang II including polyuria and hydronephrosis. The hypotensive effect of bilateral renal denervation was blunted in Agt-KO compared to wildtype FVB/N mice. CONCLUSION AND IMPLICATIONS We describe a germline Agt-KO line that challenges all previous knowledge on BP regulation in mice with deletion of the classical RAS. This line may represent a model of drug-resistant hypertension because it lacks hypotension.
Collapse
Affiliation(s)
- André Felipe Rodrigues
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Mihail Todiras
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Nicolae Testemițanu State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Fatimunnisa Qadri
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
2
|
Lucas C, Sauter KS, Steigert M, Mallet D, Wilmouth J, Olabe J, Plotton I, Morel Y, Aeberli D, Wagner F, Clevers H, Pandey AV, Val P, Roucher-Boulez F, Flück CE. Loss of LGR4/GPR48 causes severe neonatal salt wasting due to disrupted WNT signaling altering adrenal zonation. J Clin Invest 2023; 133:164915. [PMID: 36538378 PMCID: PMC9927937 DOI: 10.1172/jci164915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Disorders of isolated mineralocorticoid deficiency, which cause potentially life-threatening salt-wasting crisis early in life, have been associated with gene variants of aldosterone biosynthesis or resistance; however, in some patients no such variants are found. WNT/β-catenin signaling is crucial for differentiation and maintenance of the aldosterone-producing adrenal zona glomerulosa (zG). Herein, we describe a highly consanguineous family with multiple perinatal deaths and infants presenting at birth with failure to thrive, severe salt-wasting crises associated with isolated hypoaldosteronism, nail anomalies, short stature, and deafness. Whole exome sequencing revealed a homozygous splice variant in the R-SPONDIN receptor LGR4 gene (c.618-1G>C) regulating WNT signaling. The resulting transcripts affected protein function and stability and resulted in loss of Wnt/β-catenin signaling in vitro. The impact of LGR4 inactivation was analyzed by adrenal cortex-specific ablation of Lgr4, using Lgr4fl/fl mice mated with Sf1:Cre mice. Inactivation of Lgr4 within the adrenal cortex in the mouse model caused decreased WNT signaling, aberrant zonation with deficient zG, and reduced aldosterone production. Thus, human LGR4 mutations establish a direct link between LGR4 inactivation and decreased canonical WNT signaling, which results in abnormal zG differentiation and endocrine function. Therefore, variants in WNT signaling and its regulators should systematically be considered in familial hyperreninemic hypoaldosteronism.
Collapse
Affiliation(s)
- Cécily Lucas
- Laboratoire de Biochimie et Biologie Moléculaire, UM Pathologies Endocriniennes, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France.,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Clermont Auvergne, CNRS, Inserm, Génétique, Reproduction et Développement, Clermont-Ferrand, France
| | - Kay-Sara Sauter
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Inselspital, Bern University Hospital, and.,Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Michael Steigert
- Department of Pediatrics, Cantonal Hospital Graubuenden, Chur, Switzerland
| | - Delphine Mallet
- Laboratoire de Biochimie et Biologie Moléculaire, UM Pathologies Endocriniennes, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France.,Centre de Référence Maladies Rares du Développement Génital: du Fœtus à l'Adulte, Filière Maladies Rares Endocriniennes, Bron, France
| | - James Wilmouth
- Université Clermont Auvergne, CNRS, Inserm, Génétique, Reproduction et Développement, Clermont-Ferrand, France
| | - Julie Olabe
- Université Clermont Auvergne, CNRS, Inserm, Génétique, Reproduction et Développement, Clermont-Ferrand, France
| | - Ingrid Plotton
- Laboratoire de Biochimie et Biologie Moléculaire, UM Pathologies Endocriniennes, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France.,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence Maladies Rares du Développement Génital: du Fœtus à l'Adulte, Filière Maladies Rares Endocriniennes, Bron, France
| | - Yves Morel
- Laboratoire de Biochimie et Biologie Moléculaire, UM Pathologies Endocriniennes, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France.,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence Maladies Rares du Développement Génital: du Fœtus à l'Adulte, Filière Maladies Rares Endocriniennes, Bron, France
| | - Daniel Aeberli
- Department of Rheumatology and Clinical Immunology/Allergology and
| | - Franca Wagner
- University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Centre Utrecht, Utrecht, Netherlands
| | - Amit V Pandey
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Inselspital, Bern University Hospital, and.,Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Pierre Val
- Université Clermont Auvergne, CNRS, Inserm, Génétique, Reproduction et Développement, Clermont-Ferrand, France
| | - Florence Roucher-Boulez
- Laboratoire de Biochimie et Biologie Moléculaire, UM Pathologies Endocriniennes, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France.,University of Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Clermont Auvergne, CNRS, Inserm, Génétique, Reproduction et Développement, Clermont-Ferrand, France
| | - Christa E Flück
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Inselspital, Bern University Hospital, and.,Department of Biomedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
3
|
Qin L, Liu N, Bao CLM, Yang DZ, Ma GX, Yi WH, Xiao GZ, Cao HL. Mesenchymal stem cells in fibrotic diseases-the two sides of the same coin. Acta Pharmacol Sin 2023; 44:268-287. [PMID: 35896695 PMCID: PMC9326421 DOI: 10.1038/s41401-022-00952-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 06/29/2022] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is caused by extensive deposition of extracellular matrix (ECM) components, which play a crucial role in injury repair. Fibrosis attributes to ~45% of all deaths worldwide. The molecular pathology of different fibrotic diseases varies, and a number of bioactive factors are involved in the pathogenic process. Mesenchymal stem cells (MSCs) are a type of multipotent stem cells that have promising therapeutic effects in the treatment of different diseases. Current updates of fibrotic pathogenesis reveal that residential MSCs may differentiate into myofibroblasts which lead to the fibrosis development. However, preclinical and clinical trials with autologous or allogeneic MSCs infusion demonstrate that MSCs can relieve the fibrotic diseases by modulating inflammation, regenerating damaged tissues, remodeling the ECMs, and modulating the death of stressed cells after implantation. A variety of animal models were developed to study the mechanisms behind different fibrotic tissues and test the preclinical efficacy of MSC therapy in these diseases. Furthermore, MSCs have been used for treating liver cirrhosis and pulmonary fibrosis patients in several clinical trials, leading to satisfactory clinical efficacy without severe adverse events. This review discusses the two opposite roles of residential MSCs and external MSCs in fibrotic diseases, and summarizes the current perspective of therapeutic mechanism of MSCs in fibrosis, through both laboratory study and clinical trials.
Collapse
Affiliation(s)
- Lei Qin
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Nian Liu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Chao-le-meng Bao
- CASTD Regengeek (Shenzhen) Medical Technology Co. Ltd, Shenzhen, 518000 China
| | - Da-zhi Yang
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Gui-xing Ma
- grid.263817.90000 0004 1773 1790Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055 China
| | - Wei-hong Yi
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Guo-zhi Xiao
- grid.263817.90000 0004 1773 1790Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055 China
| | - Hui-ling Cao
- grid.263817.90000 0004 1773 1790Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055 China
| |
Collapse
|
4
|
Takayama S, Inoue K, Ogura Y, Hoshino S, Sugaya T, Ohata K, Kotake H, Ichikawa D, Watanabe M, Kimura K, Shibagaki Y, Kamijo-Ikemori A. Angiotensin II type 1a receptor deficiency alleviates muscle atrophy after denervation. Sci Rep 2023; 13:519. [PMID: 36627369 PMCID: PMC9832142 DOI: 10.1038/s41598-023-27737-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
The study aim was to determine if suppressed activation of angiotensin II type 1 receptor (AT1) prevents severe muscle atrophy after denervation. The sciatic nerves in right and left inferior limbs were cut in AT1a knockout homo (AT1a-/-) male mice and wild-type (AT1a+/+) male mice. Muscle weight and cross-sectional areas of type IIb muscle fibers in gastrocnemius muscle decreased at 7 and 21 days postdenervation in both AT1a-/- mice and AT1a+/+ mice, and the reduction was significantly attenuated in the denervated muscles of AT1a-/- mice compared to the AT1a+/+ mice. Gene expressions in the protein degradation system [two E3 ubiquitin ligases (muscle RING-finger protein-1 and Atrogin-1)] upregulated at 7 days postdenervation in all denervated mice were significantly lower in AT1a-/- mice than in AT1a+/+ mice. Activations of nuclear factor κB and Forkhead box subgroup O1, and protein expression of monocyte chemoattractant protein-1 were significantly suppressed in the AT1a-/- mice compared with those in the AT1a+/+ mice. In addition, suppressed apoptosis, lower infiltration of M1 macrophages, and higher infiltration of M2 macrophages were significantly observed at 21 days postdenervation in the AT1a-/- mice compared with those in the AT1a+/+ mice. In conclusion, the AT1 receptor deficiency retarded muscle atrophy after denervation.
Collapse
Affiliation(s)
- Suguru Takayama
- grid.412764.20000 0004 0372 3116Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-Ku, Kawasaki, 216-8511 Japan
| | - Kazuho Inoue
- grid.412764.20000 0004 0372 3116Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Yuji Ogura
- grid.412764.20000 0004 0372 3116Department of Physiology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Seiko Hoshino
- grid.412764.20000 0004 0372 3116Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Takeshi Sugaya
- grid.412764.20000 0004 0372 3116Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-Ku, Kawasaki, 216-8511 Japan
| | - Keiichi Ohata
- grid.412764.20000 0004 0372 3116Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-Ku, Kawasaki, 216-8511 Japan
| | - Hitoshi Kotake
- grid.412764.20000 0004 0372 3116Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-Ku, Kawasaki, 216-8511 Japan
| | - Daisuke Ichikawa
- grid.412764.20000 0004 0372 3116Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-Ku, Kawasaki, 216-8511 Japan
| | - Minoru Watanabe
- Institute for Animal Experimentation, St. Marianna University Graduate School of Medicine, Kanagawa, Japan
| | | | - Yugo Shibagaki
- grid.412764.20000 0004 0372 3116Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-Ku, Kawasaki, 216-8511 Japan
| | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-Ku, Kawasaki, 216-8511, Japan. .,Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan. .,Institute for Animal Experimentation, St. Marianna University Graduate School of Medicine, Kanagawa, Japan.
| |
Collapse
|
5
|
Egan BM, Scharf A, Pohl F, Kornfeld K. Control of aging by the renin–angiotensin system: a review of C. elegans, Drosophila, and mammals. Front Pharmacol 2022; 13:938650. [PMID: 36188619 PMCID: PMC9518657 DOI: 10.3389/fphar.2022.938650] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
The free-living, non-parasitic nematode Caenorhabditis elegans is a premier model organism for the study of aging and longevity due to its short lifespan, powerful genetic tools, and conservation of fundamental mechanisms with mammals. Approximately 70 percent of human genes have homologs in C. elegans, including many that encode proteins in pathways that influence aging. Numerous genetic pathways have been identified in C. elegans that affect lifespan, including the dietary restriction pathway, the insulin/insulin-like growth factor (IGF) signaling pathway, and the disruption of components of the mitochondrial electron transport chain. C. elegans is also a powerful system for performing drug screens, and many lifespan-extending compounds have been reported; notably, several FDA-approved medications extend the lifespan in C. elegans, raising the possibility that they can also extend the lifespan in humans. The renin–angiotensin system (RAS) in mammals is an endocrine system that regulates blood pressure and a paracrine system that acts in a wide range of tissues to control physiological processes; it is a popular target for drugs that reduce blood pressure, including angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs). Emerging evidence indicates that this system influences aging. In C. elegans, decreasing the activity of the ACE homolog acn-1 or treatment with the ACE-inhibitor Captopril significantly extends the lifespan. In Drosophila, treatment with ACE inhibitors extends the lifespan. In rodents, manipulating the RAS with genetic or pharmacological interventions can extend the lifespan. In humans, polymorphisms in the ACE gene are associated with extreme longevity. These results suggest the RAS plays a conserved role in controlling longevity. Here, we review studies of the RAS and aging, emphasizing the potential of C. elegans as a model for understanding the mechanism of lifespan control.
Collapse
Affiliation(s)
- Brian M. Egan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
| | - Andrea Scharf
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, United States
| | - Franziska Pohl
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Kerry Kornfeld
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
- *Correspondence: Kerry Kornfeld,
| |
Collapse
|
6
|
Angiotensin II type 1a receptor loss ameliorates chronic tubulointerstitial damage after renal ischemia reperfusion. Sci Rep 2021; 11:982. [PMID: 33441837 PMCID: PMC7806698 DOI: 10.1038/s41598-020-80209-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 12/14/2020] [Indexed: 12/26/2022] Open
Abstract
We investigate whether suppressing the activation of the angiotensin II type 1a receptor (AT1a) can ameliorate severe chronic tubulointerstitial damage (TID) after renal ischemia reperfusion (IR) using AT1a knockout homozygous (AT1a−/−) male mice. To induce severe chronic TID after renal IR, unilateral renal ischemia was performed via clamping of the right renal pedicle in both AT1a−/− and wild-type (AT1a+/+) mice for 45 min. While marked renal atrophy and severe TID at 70 days postischemia was induced in the AT1a+/+ mice, such a development was not provoked in the AT1a−/− mice. Although the AT1a+/+ mice were administered hydralazine to maintain the same systolic blood pressure (SBP) levels as the AT1a−/− mice with lower SBP levels, hydralazine did not reproduce the renoprotective effects observed in the AT1a−/− mice. Acute tubular injury at 3 days postischemia was similar between the AT1a−/− mice and the AT1a+/+ mice. From our investigations using IR kidneys at 3, 14, and 28 days postischemia, the multiple molecular mechanisms may be related to prevention of severe chronic TID postischemia in the AT1a−/− mice. In conclusion, inactivation of the AT1 receptor may be useful in preventing the transition of acute kidney injury to chronic kidney disease.
Collapse
|
7
|
Ji X, Tan W, Zhang C, Zhai Y, Hsueh Y, Zhang Z, Zhang C, Lu Y, Duan B, Tan G, Na R, Deng G, Niu G. TWIRLS, a knowledge-mining technology, suggests a possible mechanism for the pathological changes in the human host after coronavirus infection via ACE2. Drug Dev Res 2020; 81:1004-1018. [PMID: 32657473 PMCID: PMC7404951 DOI: 10.1002/ddr.21717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/05/2020] [Accepted: 06/27/2020] [Indexed: 12/12/2022]
Abstract
Faced with the current large-scale public health emergency, collecting, sorting, and analyzing biomedical information related to the "SARS-CoV-2" should be done as quickly as possible to gain a global perspective, which is a basic requirement for strengthening epidemic control capacity. However, for human researchers studying viruses and hosts, the vast amount of information available cannot be processed effectively and in a timely manner, particularly if our scientific understanding is also limited, which further lowers the information processing efficiency. We present TWIRLS (Topic-wise inference engine of massive biomedical literatures), a method that can deal with various scientific problems, such as liver cancer, acute myeloid leukemia, and so forth, which can automatically acquire, organize, and classify information. Additionally, this information can be combined with independent functional data sources to build an inference system via a machine-based approach, which can provide relevant knowledge to help human researchers quickly establish subject cognition and to make more effective decisions. Using TWIRLS, we automatically analyzed more than three million words in more than 14,000 literature articles in only 4 hr. We found that an important regulatory factor angiotensin-converting enzyme 2 (ACE2) may be involved in host pathological changes on binding to the coronavirus after infection. On triggering functional changes in ACE2/AT2R, the cytokine homeostasis regulation axis becomes imbalanced via the Renin-Angiotensin System and IP-10, leading to a cytokine storm. Through a preliminary analysis of blood indices of COVID-19 patients with a history of hypertension, we found that non-ARB (Angiotensin II receptor blockers) users had more symptoms of severe illness than ARB users. This suggests ARBs could potentially be used to treat acute lung injury caused by coronavirus infection.
Collapse
Affiliation(s)
- Xiaoyang Ji
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Inner Mongolia Autonomous Region College of Animal ScienceInner Mongolia Agricultural UniversityHohhotChina
- Joint Turing‐Darwin Laboratory of Phil Rivers Technology Ltd. and Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- Department of Computational Biology, Phil Rivers Technology LtdBeijingChina
| | - Wenting Tan
- Department of Infectious DiseasesSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Chunming Zhang
- Joint Turing‐Darwin Laboratory of Phil Rivers Technology Ltd. and Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- Department of Computational Biology, Phil Rivers Technology LtdBeijingChina
- Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- West Institute of Computing TechnologyChinese Academy of SciencesChongqingChina
| | - Yubo Zhai
- Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yiching Hsueh
- Joint Turing‐Darwin Laboratory of Phil Rivers Technology Ltd. and Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- Department of Computational Biology, Phil Rivers Technology LtdBeijingChina
| | - Zhonghai Zhang
- Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
| | - Chunli Zhang
- Department of Computational Biology, Phil Rivers Technology LtdBeijingChina
| | - Yanqiu Lu
- Department of Infectious DiseasesChongqing Public Health Medical CenterChongqingChina
| | - Bo Duan
- Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- West Institute of Computing TechnologyChinese Academy of SciencesChongqingChina
| | - Guangming Tan
- Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- West Institute of Computing TechnologyChinese Academy of SciencesChongqingChina
| | - Renhua Na
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Inner Mongolia Autonomous Region College of Animal ScienceInner Mongolia Agricultural UniversityHohhotChina
| | - Guohong Deng
- Department of Infectious DiseasesSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingChina
| | - Gang Niu
- Joint Turing‐Darwin Laboratory of Phil Rivers Technology Ltd. and Institute of Computing TechnologyChinese Academy of SciencesBeijingChina
- Department of Computational Biology, Phil Rivers Technology LtdBeijingChina
- West Institute of Computing TechnologyChinese Academy of SciencesChongqingChina
| |
Collapse
|
8
|
Kim M, Do GY, Kim I. Activation of the renin-angiotensin system in high fructose-induced metabolic syndrome. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:319-328. [PMID: 32587126 PMCID: PMC7317175 DOI: 10.4196/kjpp.2020.24.4.319] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 01/02/2023]
Abstract
High fructose intake induces hyperglycemia and hypertension. However, the mechanism by which fructose induces metabolic syndrome is largely unknown. We hypothesized that high fructose intake induces activation of the renin-angiotensin system (RAS), resulting in hypertension and metabolic syndrome. We provided 11-week-old Sprague–Dawley rats with drinking water, with or without 20% fructose, for two weeks. We measured serum renin, angiotensin II (Ang II), and aldosterone (Aldo) using ELISA kits. The expression of RAS genes was determined by quantitative reverse transcription polymerase chain reaction. High fructose intake increased body weight and water retention, regardless of food intake or urine volume. After two weeks, fructose intake induced glucose intolerance and hypertension. High fructose intake increased serum renin, Ang II, triglyceride, and cholesterol levels, but not Aldo levels. High fructose intake increased the expression of angiotensinogen in the liver; angiotensin-converting enzyme in the lungs; and renin, angiotensin II type 1a receptor (AT1aR), and angiotensin II type 1b receptor (AT1bR) in the kidneys. However, expression of AT1aR and AT1bR in the adrenal glands did not increase in rats given fructose. Taken together, these results indicate that high fructose intake induces activation of RAS, resulting in hypertension and metabolic syndrome.
Collapse
Affiliation(s)
- Mina Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Ga Young Do
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Inkyeom Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,Cardiovascular Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
9
|
Sakuta H, Lin CH, Hiyama TY, Matsuda T, Yamaguchi K, Shigenobu S, Kobayashi K, Noda M. SLC9A4 in the organum vasculosum of the lamina terminalis is a [Na+] sensor for the control of water intake. Pflugers Arch 2020; 472:609-624. [DOI: 10.1007/s00424-020-02389-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/17/2020] [Accepted: 04/28/2020] [Indexed: 12/19/2022]
|
10
|
Otani K, Tokudome T, Kamiya CA, Mao Y, Nishimura H, Hasegawa T, Arai Y, Kaneko M, Shioi G, Ishida J, Fukamizu A, Osaki T, Nagai-Okatani C, Minamino N, Ensho T, Hino J, Murata S, Takegami M, Nishimura K, Kishimoto I, Miyazato M, Harada-Shiba M, Yoshimatsu J, Nakao K, Ikeda T, Kangawa K. Deficiency of Cardiac Natriuretic Peptide Signaling Promotes Peripartum Cardiomyopathy-Like Remodeling in the Mouse Heart. Circulation 2019; 141:571-588. [PMID: 31665900 DOI: 10.1161/circulationaha.119.039761] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The maternal circulatory system and hormone balance both change dynamically during pregnancy, delivery, and the postpartum period. Although atrial natriuretic peptides and brain natriuretic peptides produced in the heart control circulatory homeostasis through their common receptor, NPR1, the physiologic and pathophysiologic roles of endogenous atrial natriuretic peptide/brain natriuretic peptide in the perinatal period are not fully understood. METHODS To clarify the physiologic and pathophysiologic roles of the endogenous atrial natriuretic peptide/brain natriuretic peptide-NPR1 system during the perinatal period, the phenotype of female wild-type and conventional or tissue-specific Npr1-knockout mice during the perinatal period was examined, especially focusing on maternal heart weight, blood pressure, and cardiac function. RESULTS In wild-type mice, lactation but not pregnancy induced reversible cardiac hypertrophy accompanied by increases in fetal cardiac gene mRNAs and ERK1/2 (extracellular signaling-regulated kinase) phosphorylation. Npr1-knockout mice exhibited significantly higher plasma aldosterone level than did wild-type mice, severe cardiac hypertrophy accompanied by fibrosis, and left ventricular dysfunction in the lactation period. Npr1-knockout mice showed a high mortality rate over consecutive pregnancy-lactation cycles. In the hearts of Npr1-knockout mice during or after the lactation period, an increase in interleukin-6 mRNA expression, phosphorylation of signal transducer and activator of transcription 3, and activation of the calcineurin-nuclear factor of the activated T cells pathway were observed. Pharmacologic inhibition of the mineralocorticoid receptor or neuron-specific deletion of the mineralocorticoid receptor gene significantly ameliorated cardiac hypertrophy in lactating Npr1-knockout mice. Anti-interleukin-6 receptor antibody administration tended to reduce cardiac hypertrophy in lactating Npr1-knockout mice. CONCLUSIONS These results suggest that the characteristics of lactation-induced cardiac hypertrophy in wild-type mice are different from exercise-induced cardiac hypertrophy, and that the endogenous atrial natriuretic peptide/brain natriuretic peptide-NPR1 system plays an important role in protecting the maternal heart from interleukin-6-induced inflammation and remodeling in the lactation period, a condition mimicking peripartum cardiomyopathy.
Collapse
Affiliation(s)
- Kentaro Otani
- Departments of Regenerative Medicine and Tissue Engineering (K.O., M.H.-S., T.I.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Takeshi Tokudome
- Biochemistry (T.T., Y.M., H.N., T.E., J.H., I.K., M.M.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Chizuko A Kamiya
- Division of Perinatology and Gynecology (C.A.K., J.Y.), Osaka, Japan
| | - Yuanjie Mao
- Biochemistry (T.T., Y.M., H.N., T.E., J.H., I.K., M.M.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan.,Diabetes Institute, Ohio University, Athens (Y.M.)
| | - Hirohito Nishimura
- Biochemistry (T.T., Y.M., H.N., T.E., J.H., I.K., M.M.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Takeshi Hasegawa
- Exploratory Research Section II, Exploratory Research Laboratories, TOA EIYO Ltd, Fukushima, Japan (T.H.)
| | - Yuji Arai
- Bioscience and Genetics (Y.A.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Mari Kaneko
- Animal Resource Development Unit (M.K.), RIKEN Center for Life Science Technologies, Hyogo, Japan.,Genetic Engineering Team (M.K., G.S.), RIKEN Center for Life Science Technologies, Hyogo, Japan
| | - Go Shioi
- Genetic Engineering Team (M.K., G.S.), RIKEN Center for Life Science Technologies, Hyogo, Japan
| | - Junji Ishida
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki, Japan (J.I., A.F.)
| | - Akiyoshi Fukamizu
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki, Japan (J.I., A.F.)
| | - Tsukasa Osaki
- Molecular Pharmacology (T.O., C.N.-O., N.M.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Chiaki Nagai-Okatani
- Molecular Pharmacology (T.O., C.N.-O., N.M.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Naoto Minamino
- Molecular Pharmacology (T.O., C.N.-O., N.M.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Takuya Ensho
- Biochemistry (T.T., Y.M., H.N., T.E., J.H., I.K., M.M.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Jun Hino
- Biochemistry (T.T., Y.M., H.N., T.E., J.H., I.K., M.M.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Shunsuke Murata
- Preventive Medicine and Epidemiology (S.M., M.T., K.N.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Misa Takegami
- Preventive Medicine and Epidemiology (S.M., M.T., K.N.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Kunihiro Nishimura
- Preventive Medicine and Epidemiology (S.M., M.T., K.N.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Ichiro Kishimoto
- Biochemistry (T.T., Y.M., H.N., T.E., J.H., I.K., M.M.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Mikiya Miyazato
- Biochemistry (T.T., Y.M., H.N., T.E., J.H., I.K., M.M.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Mariko Harada-Shiba
- Departments of Regenerative Medicine and Tissue Engineering (K.O., M.H.-S., T.I.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan.,Molecular Innovation in Lipidology (M.H.-S.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Jun Yoshimatsu
- Division of Perinatology and Gynecology (C.A.K., J.Y.), Osaka, Japan
| | - Kazuwa Nakao
- Kyoto University Graduate School of Medicine Medical Innovation Center, Kyoto, Japan (K.N.)
| | - Tomoaki Ikeda
- Departments of Regenerative Medicine and Tissue Engineering (K.O., M.H.-S., T.I.), National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan.,Department of Obstetrics and Gynecology, Mie University Graduate School of Medicine, Japan (T.I.)
| | - Kenji Kangawa
- National Cerebral and Cardiovascular Center (K.K.), Osaka, Japan
| |
Collapse
|
11
|
Takimoto-Ohnishi E, Murakami K. Renin-angiotensin system research: from molecules to the whole body. J Physiol Sci 2019; 69:581-587. [PMID: 31028527 PMCID: PMC10717639 DOI: 10.1007/s12576-019-00679-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 04/17/2019] [Indexed: 10/26/2022]
Abstract
Hypertension is one of the most important risk factors and a leading cause of death from cardiovascular and cerebrovascular diseases. Based on numerous previous studies, hypertension is thought to be caused by the complex mutual interactions of genetic factors and environmental factors, such as excessive salt intake and stress. However, its detailed mechanisms are not yet clearly understood. The renin-angiotensin system (RAS) is a key hormonal system in the pathogenesis of hypertension. New knowledge is still accruing on this cascade, even after more than 120 years since the discovery of renin. To clarify the molecular mechanisms of RAS in vivo, we created transgenic mice with chronic hypertension. These mice carry the human genes encoding renin, a hypertensive enzyme, and its substrate angiotensinogen. Hypotensive mice homozygous for a targeted disruption of the angiotensinogen gene were also created. This review presents our 47-year history of RAS research.
Collapse
Affiliation(s)
- Eriko Takimoto-Ohnishi
- Bio-Laboratory, Foundation for Advancement of International Science, 3-24-16 Kasuga, Tsukuba, Ibaraki, 305-0821, Japan.
- Department of Maternal-Fetal Biology, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan.
| | - Kazuo Murakami
- Bio-Laboratory, Foundation for Advancement of International Science, 3-24-16 Kasuga, Tsukuba, Ibaraki, 305-0821, Japan.
| |
Collapse
|
12
|
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: 104] [Impact Index Per Article: 17.3] [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
|
13
|
Neubauer B, Schrankl J, Steppan D, Neubauer K, Sequeira-Lopez ML, Pan L, Gomez RA, Coffman TM, Gross KW, Kurtz A, Wagner C. Angiotensin II Short-Loop Feedback: Is There a Role of Ang II for the Regulation of the Renin System In Vivo? Hypertension 2018; 71:1075-1082. [PMID: 29661841 DOI: 10.1161/hypertensionaha.117.10357] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/11/2017] [Accepted: 03/19/2018] [Indexed: 11/16/2022]
Abstract
The activity of the renin-angiotensin-aldosterone system is triggered by the release of the protease renin from the kidneys, which in turn is controlled in the sense of negative feedback loops. It is widely assumed that Ang II (angiotensin II) directly inhibits renin expression and secretion via a short-loop feedback by an effect on renin-producing cells (RPCs) mediated by AT1 (Ang II type 1) receptors. Because the concept of such a direct short-loop negative feedback control, which originates mostly from in vitro experiments, has not yet been systematically proven in vivo, we aimed to test the validity of this concept by studying the regulation of renin synthesis and secretion in mice lacking Ang II-AT1 receptors on RPCs. We found that RPCs of the kidney express Ang II-AT1 receptors. Mice with conditional deletion of Ang II-AT1 receptors in RPCs were normal with regard to the number of renin cells, renal renin mRNA, and plasma renin concentrations. Renin expression and secretion of these mice responded to Ang I (angiotensin I)-converting enzyme inhibition and to Ang II infusion like in wild-type (WT) controls. In summary, we did not obtain evidence that Ang II-AT1 receptors on RPCs are of major relevance for the normal regulation of renin expression and secretion in mice. Therefore, we doubt the existence of a direct negative feedback function of Ang II on RPCs.
Collapse
Affiliation(s)
- Bjoern Neubauer
- From the Institute of Physiology, University of Regensburg, Germany (B.N., J.S., D.S., K.N., A.K., C.W.)
| | - Julia Schrankl
- From the Institute of Physiology, University of Regensburg, Germany (B.N., J.S., D.S., K.N., A.K., C.W.)
| | - Dominik Steppan
- From the Institute of Physiology, University of Regensburg, Germany (B.N., J.S., D.S., K.N., A.K., C.W.)
| | - Katharina Neubauer
- From the Institute of Physiology, University of Regensburg, Germany (B.N., J.S., D.S., K.N., A.K., C.W.).,Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Germany (K.N.)
| | - Maria Luisa Sequeira-Lopez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville (M.L.S.-L., R.A.G.)
| | - Li Pan
- Department of Pathology, Brigham and Women's Hospital, Boston, MA (L.P.)
| | - R Ariel Gomez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville (M.L.S.-L., R.A.G.)
| | - Thomas M Coffman
- Division of Nephrology, Department of Medicine, Durham Veterans Affairs Medical Centers, Duke University, NC (T.M.C.).,and Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY (K.W.G.)
| | - Kenneth W Gross
- and Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY (K.W.G.)
| | - Armin Kurtz
- From the Institute of Physiology, University of Regensburg, Germany (B.N., J.S., D.S., K.N., A.K., C.W.)
| | - Charlotte Wagner
- From the Institute of Physiology, University of Regensburg, Germany (B.N., J.S., D.S., K.N., A.K., C.W.)
| |
Collapse
|
14
|
COX-2-derived PGE 2 triggers hyperplastic renin expression and hyperreninemia in aldosterone synthase-deficient mice. Pflugers Arch 2018; 470:1127-1137. [PMID: 29455241 PMCID: PMC6013527 DOI: 10.1007/s00424-018-2118-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/26/2018] [Accepted: 02/01/2018] [Indexed: 11/25/2022]
Abstract
Pharmacological inhibition or genetic loss of function defects of the renin angiotensin aldosterone system (RAAS) causes compensatory renin cell hyperplasia and hyperreninemia. The triggers for the compensatory stimulation of renin synthesis and secretion in this situation may be multimodal. Since cyclooxygenase-2 (COX-2) expression in the macula densa is frequently increased in states of a defective RAAS, we have investigated a potential role of COX-2 and its derived prostaglandins for renin expression and secretion in aldosterone synthase-deficient mice (AS−/−) as a model for a genetic defect of the RAAS. In comparison with wild-type mice (WT), AS−/− mice had 9-fold and 30-fold increases of renin mRNA and of plasma renin concentrations (PRC), respectively. Renin immunoreactivity in the kidney cortex of AS−/− mice was 10-fold higher than in WT. Macula densa COX-2 expression was 5-fold increased in AS−/− kidneys relative to WT kidneys. Treatment of AS−/− mice with the COX-2 inhibitor SC-236 for 1 week lowered both renal renin mRNA and PRC by 70%. Hyperplastic renin cells in AS−/− kidneys were found to express the prostaglandin E2 receptors EP2 and EP4. Global deletion of EP2 receptors did not alter renin mRNA nor PRC values in AS−/− mice. Renin cell-specific inducible deletion of the EP4 receptor lowered renin mRNA and PRC by 25% in AS−/− mice. Renin cell-specific inducible deletion of the EP4 receptor in combination with global deletion of the EP2 receptor lowered renin mRNA and PRC by 70–75% in AS−/− mice. Lineage tracing of renin-expressing cells revealed that deletion of EP2 and EP4 leads to a preferential downregulation of perivascular renin expression. Our findings suggest that increased macula densa COX-2 activity in AS−/− mice triggers perivascular renin expression and secretion via prostaglandin E2.
Collapse
|
15
|
Rahbari NN, Kedrin D, Incio J, Liu H, Ho WW, Nia HT, Edrich CM, Jung K, Daubriac J, Chen I, Heishi T, Martin JD, Huang Y, Maimon N, Reissfelder C, Weitz J, Boucher Y, Clark JW, Grodzinsky AJ, Duda DG, Jain RK, Fukumura D. Anti-VEGF therapy induces ECM remodeling and mechanical barriers to therapy in colorectal cancer liver metastases. Sci Transl Med 2017; 8:360ra135. [PMID: 27733559 DOI: 10.1126/scitranslmed.aaf5219] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 09/13/2016] [Indexed: 12/17/2022]
Abstract
The survival benefit of anti-vascular endothelial growth factor (VEGF) therapy in metastatic colorectal cancer (mCRC) patients is limited to a few months because of acquired resistance. We show that anti-VEGF therapy induced remodeling of the extracellular matrix with subsequent alteration of the physical properties of colorectal liver metastases. Preoperative treatment with bevacizumab in patients with colorectal liver metastases increased hyaluronic acid (HA) deposition within the tumors. Moreover, in two syngeneic mouse models of CRC metastasis in the liver, we show that anti-VEGF therapy markedly increased the expression of HA and sulfated glycosaminoglycans (sGAGs), without significantly changing collagen deposition. The density of these matrix components correlated with increased tumor stiffness after anti-VEGF therapy. Treatment-induced tumor hypoxia appeared to be the driving force for the remodeling of the extracellular matrix. In preclinical models, we show that enzymatic depletion of HA partially rescued the compromised perfusion in liver mCRCs after anti-VEGF therapy and prolonged survival in combination with anti-VEGF therapy and chemotherapy. These findings suggest that extracellular matrix components such as HA could be a potential therapeutic target for reducing physical barriers to systemic treatments in patients with mCRC who receive anti-VEGF therapy.
Collapse
Affiliation(s)
- Nuh N Rahbari
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. Department of General, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Dmitriy Kedrin
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Joao Incio
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hao Liu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - William W Ho
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hadi T Nia
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Christina M Edrich
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Keehoon Jung
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Julien Daubriac
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Ivy Chen
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. Harvard School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Takahiro Heishi
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - John D Martin
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yuhui Huang
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Nir Maimon
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Christoph Reissfelder
- Department of General, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Jurgen Weitz
- Department of General, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Yves Boucher
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jeffrey W Clark
- Department of Hematology/Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Alan J Grodzinsky
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dan G Duda
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Dai Fukumura
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
16
|
Stilhano RS, Samoto VY, Silva LM, Pereira GJ, Erustes AG, Smaili SS, Won Han S. Reduction in skeletal muscle fibrosis of spontaneously hypertensive rats after laceration by microRNA targeting angiotensin II receptor. PLoS One 2017; 12:e0186719. [PMID: 29059221 PMCID: PMC5653346 DOI: 10.1371/journal.pone.0186719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 10/08/2017] [Indexed: 12/23/2022] Open
Abstract
Regeneration of injured skeletal muscles is affected by fibrosis, which can be improved by the administration of angiotensin II (AngII) receptor (ATR) blockers in normotensive animals. However, the role of ATR in skeletal muscle fibrosis in hypertensive organisms has not been investigated yet. The tibialis anterior (TA) muscle of spontaneously hypertensive (SHR) and Wistar rats (WR) were lacerated and a lentivector encoding a microRNA targeting AngII receptor type 1 (At1) (Lv-mirAT1a) or control (Lv-mirCTL) was injected. The TA muscles were collected after 30 days to evaluate fibrosis by histology and gene expression by real-time quantitative PCR (RT-qPCR) and Western blot. SHR's myoblasts were analyzed by RT-qPCR, 48 h after transduction. In the SHR's TA, AT1 protein expression was 23.5-fold higher than in WR without injury, but no difference was observed in the angiotensin II receptor type 2 (AT2) protein expression. TA laceration followed by suture (LS) produced fibrosis in the SHR (23.3±8.5%) and WR (7.9±1.5%). Lv-mirAT1 treatment decreased At1 gene expression in 50% and reduced fibrosis to 7% 30 days after. RT-qPCR showed that reduction in At1 expression is due to downregulation of the At1a but not of the At1b. RT-qPCR of myoblasts from SHR transduced with Lv-mirAT1a showed downregulation of the Tgf-b1, Tgf-b2, Smad3, Col1a1, and Col3a1 genes by mirAT1a. In vivo and in vitro studies indicate that hypertension overproduces skeletal muscle fibrosis, and AngII-AT1a signaling is the main pathway of fibrosis in SHR. Moreover, muscle fibrosis can be treated specifically by in loco injection of Lv-mirAT1a without affecting other organs.
Collapse
Affiliation(s)
- Roberta Sessa Stilhano
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Vivian Yochiko Samoto
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Leonardo Martins Silva
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Gustavo José Pereira
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Adolfo Garcia Erustes
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Soraya Soubhi Smaili
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Sang Won Han
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
- * E-mail:
| |
Collapse
|
17
|
Zimnol A, Amann K, Mandel P, Hartmann C, Schupp N. Angiotensin II type 1a receptor-deficient mice develop angiotensin II-induced oxidative stress and DNA damage without blood pressure increase. Am J Physiol Renal Physiol 2017; 313:F1264-F1273. [PMID: 28877878 DOI: 10.1152/ajprenal.00183.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 08/04/2017] [Accepted: 08/29/2017] [Indexed: 11/22/2022] Open
Abstract
Hypertensive patients have an increased risk of developing kidney cancer. We have shown in vivo that besides elevating blood pressure, angiotensin II causes DNA damage dose dependently. Here, the role of blood pressure in the formation of DNA damage is studied. Mice lacking one of the two murine angiotensin II type 1 receptor (AT1R) subtypes, AT1aR, were equipped with osmotic minipumps, delivering angiotensin II during 28 days. Parameters of oxidative stress and DNA damage of kidneys and hearts of AT1aR-knockout mice were compared with wild-type (C57BL/6) mice receiving angiotensin II, and additionally, with wild-type mice treated with candesartan, an antagonist of both AT1R subtypes. In wild-type mice, angiotensin II induced hypertension, reduced kidney function, and led to a significant formation of reactive oxygen species (ROS). Furthermore, genomic damage was markedly increased in this group. All these responses to angiotensin II could be attenuated by concurrent administration of candesartan. In AT1aR-deficient mice treated with angiotensin II, systolic pressure was not increased, and renal function was not affected. However, angiotensin II still led to an increase of ROS in kidneys and hearts of these animals. Additionally, genomic damage in the form of double-strand breaks was significantly induced in kidneys of AT1aR-deficient mice. Our results show that angiotensin II induced ROS production and DNA damage even without the presence of AT1aR and independently of blood pressure changes.
Collapse
Affiliation(s)
- Anna Zimnol
- Institute of Toxicology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany; and
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Mandel
- Institute of Toxicology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany; and
| | - Christina Hartmann
- Institute of Toxicology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany; and
| | - Nicole Schupp
- Institute of Toxicology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany; and
| |
Collapse
|
18
|
Ohata K, Kamijo-Ikemori A, Sugaya T, Hibi C, Nakamura T, Murase T, Oikawa T, Hoshino S, Katayama K, Asano J, Kimura K, Shibagaki Y. Renoprotective effect of the xanthine oxidoreductase inhibitor Topiroxostat under decreased angiotensin II type 1 a receptor expression. Eur J Pharmacol 2017; 815:88-97. [PMID: 28888756 DOI: 10.1016/j.ejphar.2017.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/02/2017] [Accepted: 09/04/2017] [Indexed: 01/13/2023]
Abstract
The aim of this study was to confirm the renoprotective effect of xanthine oxidoreductase (XOR) inhibitor, topiroxostat, compared with another XOR inhibitor, febuxostat, under decreased angiotensin II type 1a (AT1a) receptor expression in the model of renal injury caused by adenine. To evaluate the degree of tubular damage using urinary liver-type fatty acid-binding protein (L-FABP) under decreased AT1a expression, we used AT1a receptor knockdown hetero and human L-FABP chromosomal transgenic (Tg) mice (AT1a+/-L-FABP+/-). Male AT1a+/-L-FABP+/- mice were divided into two groups: the adenine diet group (n = 40) was given a diet containing only 0.2% w/w adenine, and the normal diet group (n = 5) was given a normal diet. When renal dysfunction was confirmed in the adenine diet group 4 weeks after starting the diet, the adenine diet group was further divided into five groups. The adenine diet group (n = 8) was continuously given only the adenine diet. Each group receiving high-dose (3mg/kg) or low-dose (1mg/kg) topiroxostat (Topiroxostat-H group, n = 8, Topiroxostat-L group, n = 8) or febuxostat (Febuxostat-H group, n = 8, Febuxostat-L group, n = 8) was given the adenine diet including the drug for another 4 weeks. The levels of renal XOR, renal dysfunction, urinary L-FABP, tubulointerstitial damage, hypoxia, and oxidative stress were decreased or attenuated after treatment with topiroxostat or febuxostat compared with the adenine diet group. Furthermore, antioxidant capacity was maintained owing to these treatments. In conclusion, topiroxostat and febuxostat attenuated renal damage under decreased AT1a expression in the adenine-induced renal injury model.
Collapse
Affiliation(s)
- Keiichi Ohata
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; CMIC Holdings Company, Limited, Tokyo, Japan
| | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan.
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; CMIC Holdings Company, Limited, Tokyo, Japan
| | - Chihiro Hibi
- Biopharmaceutical Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho Company, Limited, Mie, Japan
| | - Takashi Nakamura
- Biopharmaceutical Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho Company, Limited, Mie, Japan
| | - Takayo Murase
- Radioisotope and Chemical Analysis Center, Laboratory Management Department, Sanwa Kagaku Kenkyusho Company, Limited, Mie, Japan
| | | | - Seiko Hoshino
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kimie Katayama
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Junko Asano
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | | | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| |
Collapse
|
19
|
Pires PW, Ko E, Pritchard HA, Rudokas M, Yamasaki E, Earley S. The angiotensin II receptor type 1b is the primary sensor of intraluminal pressure in cerebral artery smooth muscle cells. J Physiol 2017; 595:4735-4753. [PMID: 28475214 PMCID: PMC5509855 DOI: 10.1113/jp274310] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 05/03/2017] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS The angiotensin II receptor type 1b (AT1 Rb ) is the primary sensor of intraluminal pressure in cerebral arteries. Pressure or membrane-stretch induced stimulation of AT1 Rb activates the TRPM4 channel and results in inward transient cation currents that depolarize smooth muscle cells, leading to vasoconstriction. Activation of either AT1 Ra or AT1 Rb with angiotensin II stimulates TRPM4 currents in cerebral artery myocytes and vasoconstriction of cerebral arteries. The expression of AT1 Rb mRNA is ∼30-fold higher than AT1 Ra in whole cerebral arteries and ∼45-fold higher in isolated cerebral artery smooth muscle cells. Higher levels of expression are likely to account for the obligatory role of AT1 Rb for pressure-induced vasoconstriction. ABSTRACT: Myogenic vasoconstriction, which reflects the intrinsic ability of smooth muscle cells to contract in response to increases in intraluminal pressure, is critically important for the autoregulation of blood flow. In smooth muscle cells from cerebral arteries, increasing intraluminal pressure engages a signalling cascade that stimulates cation influx through transient receptor potential (TRP) melastatin 4 (TRPM4) channels to cause membrane depolarization and vasoconstriction. Substantial evidence indicates that the angiotensin II receptor type 1 (AT1 R) is inherently mechanosensitive and initiates this signalling pathway. Rodents express two types of AT1 R - AT1 Ra and AT1 Rb - and conflicting studies provide support for either isoform as the primary sensor of intraluminal pressure in peripheral arteries. We hypothesized that mechanical activation of AT1 Ra increases TRPM4 currents to induce myogenic constriction of cerebral arteries. However, we found that development of myogenic tone was greater in arteries from AT1 Ra knockout animals compared with controls. In patch-clamp experiments using native cerebral arterial myocytes, membrane stretch-induced cation currents were blocked by the TRPM4 inhibitor 9-phenanthrol in both groups. Further, the AT1 R blocker losartan (1 μm) diminished myogenic tone and blocked stretch-induced cation currents in cerebral arteries from both groups. Activation of AT1 R with angiotensin II (30 nm) also increased TRPM4 currents in smooth muscle cells and constricted cerebral arteries from both groups. Expression of AT1 Rb mRNA was ∼30-fold greater than AT1 Ra in cerebral arteries, and knockdown of AT1 Rb selectively diminished myogenic constriction. We conclude that AT1 Rb , acting upstream of TRPM4 channels, is the primary sensor of intraluminal pressure in cerebral artery smooth muscle cells.
Collapse
Affiliation(s)
- Paulo W. Pires
- Department of Pharmacology, Center for Cardiovascular Research, University of NevadaReno School of MedicineRenoNV89557‐0318USA
| | - Eun‐A. Ko
- Department of Physiology and Cell Biology, University of NevadaReno School of MedicineRenoNV89557‐0318USA
| | - Harry A.T. Pritchard
- Department of Pharmacology, Center for Cardiovascular Research, University of NevadaReno School of MedicineRenoNV89557‐0318USA
| | - Michael Rudokas
- Department of Pharmacology, Center for Cardiovascular Research, University of NevadaReno School of MedicineRenoNV89557‐0318USA
| | - Evan Yamasaki
- Department of Pharmacology, Center for Cardiovascular Research, University of NevadaReno School of MedicineRenoNV89557‐0318USA
| | - Scott Earley
- Department of Pharmacology, Center for Cardiovascular Research, University of NevadaReno School of MedicineRenoNV89557‐0318USA
| |
Collapse
|
20
|
Nogueira A, Pires MJ, Oliveira PA. Pathophysiological Mechanisms of Renal Fibrosis: A Review of Animal Models and Therapeutic Strategies. ACTA ACUST UNITED AC 2017; 31:1-22. [PMID: 28064215 DOI: 10.21873/invivo.11019] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 10/27/2016] [Accepted: 10/31/2016] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease (CKD) is a long-term condition in which the kidneys do not work correctly. It has a high prevalence and represents a serious hazard to human health and estimated to affects hundreds of millions of people. Diabetes and hypertension are the two principal causes of CKD. The progression of CKD is characterized by the loss of renal cells and their replacement by extracellular matrix (ECM), independently of the associated disease. Thus, one of the consequences of CKD is glomerulosclerosis and tubulointerstitial fibrosis caused by an imbalance between excessive synthesis and reduced breakdown of the ECM. There are many molecules and cells that are associated with progression of renal fibrosis e.g. angiotensin II (Ang II). Therefore, in order to understand the biopathology of renal fibrosis and for the evaluation of new treatments, the use of animal models is crucial such as: surgical, chemical and physical models, spontaneous models, genetic models and in vitro models. However, there are currently no effective treatments for preventing the progression of renal fibrosis. Therefore it is essential to improve our knowledge of the cellular and molecular mechanisms of the progress of renal fibrosis in order to achieve a reversion/elimination of renal fibrosis.
Collapse
Affiliation(s)
- António Nogueira
- Center for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Department of Therapeutic and Diagnostic Technologies, Polytechnic Institute of Bragança, Bragança, Portugal
| | - Maria João Pires
- Center for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Paula Alexandra Oliveira
- Center for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal .,Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| |
Collapse
|
21
|
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
|
22
|
Enhancement of Adipocyte Browning by Angiotensin II Type 1 Receptor Blockade. PLoS One 2016; 11:e0167704. [PMID: 27992452 PMCID: PMC5167230 DOI: 10.1371/journal.pone.0167704] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 11/20/2016] [Indexed: 01/22/2023] Open
Abstract
Browning of white adipose tissue (WAT) has been highlighted as a new possible therapeutic target for obesity, diabetes and lipid metabolic disorders, because WAT browning could increase energy expenditure and reduce adiposity. The new clusters of adipocytes that emerge with WAT browning have been named ‘beige’ or ‘brite’ adipocytes. Recent reports have indicated that the renin-angiotensin system (RAS) plays a role in various aspects of adipose tissue physiology and dysfunction. The biological effects of angiotensin II, a major component of RAS, are mediated by two receptor subtypes, angiotensin II type 1 receptor (AT1R) and type 2 receptor (AT2R). However, the functional roles of angiotensin II receptor subtypes in WAT browning have not been defined. Therefore, we examined whether deletion of angiotensin II receptor subtypes (AT1aR and AT2R) may affect white-to-beige fat conversion in vivo. AT1a receptor knockout (AT1aKO) mice exhibited increased appearance of multilocular lipid droplets and upregulation of thermogenic gene expression in inguinal white adipose tissue (iWAT) compared to wild-type (WT) mice. AT2 receptor-deleted mice did not show miniaturization of lipid droplets or alteration of thermogenic gene expression levels in iWAT. An in vitro experiment using adipose tissue-derived stem cells showed that deletion of the AT1a receptor resulted in suppression of adipocyte differentiation, with reduction in expression of thermogenic genes. These results indicate that deletion of the AT1a receptor might have some effects on the process of browning of WAT and that blockade of the AT1 receptor could be a therapeutic target for the treatment of metabolic disorders.
Collapse
|
23
|
Matsuda T, Hiyama TY, Niimura F, Matsusaka T, Fukamizu A, Kobayashi K, Kobayashi K, Noda M. Distinct neural mechanisms for the control of thirst and salt appetite in the subfornical organ. Nat Neurosci 2016; 20:230-241. [PMID: 27991901 DOI: 10.1038/nn.4463] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/17/2016] [Indexed: 12/20/2022]
Abstract
Body fluid conditions are continuously monitored in the brain to regulate thirst and salt-appetite sensations. Angiotensin II drives both thirst and salt appetite; however, the neural mechanisms underlying selective water- and/or salt-intake behaviors remain unknown. Using optogenetics, we show that thirst and salt appetite are driven by distinct groups of angiotensin II receptor type 1a-positive excitatory neurons in the subfornical organ. Neurons projecting to the organum vasculosum lamina terminalis control water intake, while those projecting to the ventral part of the bed nucleus of the stria terminalis control salt intake. Thirst-driving neurons are suppressed under sodium-depleted conditions through cholecystokinin-mediated activation of GABAergic neurons. In contrast, the salt appetite-driving neurons were suppressed under dehydrated conditions through activation of another population of GABAergic neurons by Nax signals. These distinct mechanisms in the subfornical organ may underlie the selective intakes of water and/or salt and may contribute to body fluid homeostasis.
Collapse
Affiliation(s)
- Takashi Matsuda
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Aichi, Japan.,School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Takeshi Y Hiyama
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Aichi, Japan.,School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Fumio Niimura
- Department of Pediatrics, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Taiji Matsusaka
- Department of Molecular Life Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Akiyoshi Fukamizu
- Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan.,Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Masaharu Noda
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Aichi, Japan.,School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| |
Collapse
|
24
|
Lin HY, Lee YT, Chan YW, Tse G. Animal models for the study of primary and secondary hypertension in humans. Biomed Rep 2016; 5:653-659. [PMID: 28105333 PMCID: PMC5228353 DOI: 10.3892/br.2016.784] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/02/2016] [Indexed: 12/17/2022] Open
Abstract
Hypertension is a significant cause of morbidity and mortality worldwide. It is defined as systolic and diastolic blood pressures (SBP/DBP) >140 and 90 mmHg, respectively. Individuals with an SBP between 120 and 139, or DBP between 80 and 89 mmHg, are said to exhibit pre-hypertension. Hypertension can have primary or secondary causes. Primary or essential hypertension is a multifactorial disease caused by interacting environmental and polygenic factors. Secondary causes are renovascular hypertension, renal disease, endocrine disorders and other medical conditions. The aim of the present review article was to examine the different animal models that have been generated for studying the molecular and physiological mechanisms underlying hypertension. Their advantages, disadvantages and limitations will be discussed.
Collapse
Affiliation(s)
- Hiu Yu Lin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Yee Ting Lee
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Yin Wah Chan
- School of Biological Sciences, University of Cambridge, Cambridge CB2 1AG, UK
| | - Gary Tse
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, SAR, P.R. China
| |
Collapse
|
25
|
Mederos y Schnitzler M, Storch U, Gudermann T. Mechanosensitive Gq/11Protein-Coupled Receptors Mediate Myogenic Vasoconstriction. Microcirculation 2016; 23:621-625. [DOI: 10.1111/micc.12293] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/18/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Michael Mederos y Schnitzler
- Walther Straub Institute of Pharmacology and Toxicology; Ludwig Maximilians University of Munich; Munich Germany
- DZHK (German Centre for Cardiovascular Research); Munich Heart Alliance; Munich Germany
| | - Ursula Storch
- Walther Straub Institute of Pharmacology and Toxicology; Ludwig Maximilians University of Munich; Munich Germany
- DZHK (German Centre for Cardiovascular Research); Munich Heart Alliance; Munich Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology; Ludwig Maximilians University of Munich; Munich Germany
- DZHK (German Centre for Cardiovascular Research); Munich Heart Alliance; Munich Germany
- Comprehensive Pneumology Center Munich (CPC-M); German Center for Lung Research; Munich Germany
| |
Collapse
|
26
|
Hisamichi M, Kamijo-Ikemori A, Sugaya T, Ichikawa D, Natsuki T, Hoshino S, Kimura K, Shibagaki Y. Role of angiotensin II type 1a receptor in renal injury induced by deoxycorticosterone acetate-salt hypertension. FASEB J 2016; 31:72-84. [PMID: 27663860 PMCID: PMC5161521 DOI: 10.1096/fj.201600684rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/07/2016] [Indexed: 12/19/2022]
Abstract
The aim of this study was to investigate the in vivo role of angiotensin II type 1a (AT1a) receptor in renal damage as a result of hypertension by using transgenic mice with AT1a receptor gene disruption. Transgenic mice that express human liver-type fatty acid binding protein (L-FABP) with or without disruption of the AT1a receptor gene (L-FABP+/− AT1a−/−, and L-FABP+/− AT1a+/+, respectively) were used with urinary L-FABP as an indicator of tubulointerstitial damage. Those female mice were administered subcutaneously deoxycorticosterone acetate (DOCA)–salt tablets plus drinking water that contained 1% saline for 28 d after uninephrectomy. In L-FABP+/− AT1a+/+ mice that received DOCA-salt treatment, hypertension was induced and slight expansion of glomerular area, glomerular sclerosis, and tubulointerstitial damage were observed. In L-FABP+/− AT1a−/− mice that received DOCA-salt treatment, hypertension was similarly induced and the degree of glomerular damage was significantly more severe than in L-FABP+/− AT1a+/+-DOCA mice. Urinary L-FABP levels were significantly higher in L-FABP+/− AT1a−/−-DOCA mice compared with those in L-FABP+/− AT1a+/+-DOCA mice. Hydralazine treatment significantly attenuated renal damage that was found in L-FABP+/− AT1a−/−-DOCA mice along with a reduction in blood pressure. In summary, activation of the AT1a receptor may contribute to maintenance of the glomerular structure against hypertensive renal damage.—Hisamichi, M., Kamijo-Ikemori, A., Sugaya, T., Ichikawa, D., Natsuki, T., Hoshino, S., Kimura, K., Shibagaki, Y. Role of angiotensin II type 1a receptor in renal injury induced by deoxycorticosterone acetate–salt hypertension.
Collapse
Affiliation(s)
- Mikako Hisamichi
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan; .,Department of Anatomy, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Daisuke Ichikawa
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takayuki Natsuki
- Institute for Ultrastructural Morphology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Seiko Hoshino
- Department of Anatomy, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kenjiro Kimura
- Department of Internal Medicine, Japan Community Health Care Organization, Tokyo Takanawa Hospital, Tokyo, Japan
| | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
27
|
Kai Chen, Merrill DC, Rose JC. The Importance of Angiotensin II Subtype Receptors for Blood Pressure Control During Mouse Pregnancy. Reprod Sci 2016; 14:694-704. [DOI: 10.1177/1933719107309060] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Kai Chen
- Department of Obstetrics and Gynecology Wake Forest University School of Medcine, Winston-salem North Carolina
| | - David C. Merrill
- Department of Obstetrics and Gynecology Wake Forest University School of Medcine, Winston-salem North Carolina
| | - James C. Rose
- Department of Obstetrics and Gynecology Wake Forest University School of Medcine, Winston-salem North Carolina, Department of Physiology and Pharmacology Wake Forest University School of Medicine, Winston-Salem, North Carolina,
| |
Collapse
|
28
|
Morinelli TA, Luttrell LM, Strungs EG, Ullian ME. Angiotensin II receptors and peritoneal dialysis-induced peritoneal fibrosis. Int J Biochem Cell Biol 2016; 77:240-50. [PMID: 27167177 PMCID: PMC5038354 DOI: 10.1016/j.biocel.2016.04.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 12/22/2022]
Abstract
The vasoactive hormone angiotensin II initiates its major hemodynamic effects through interaction with AT1 receptors, a member of the class of G protein-coupled receptors. Acting through its AT1R, angiotensin II regulates blood pressure and renal salt and water balance. Recent evidence points to additional pathological influences of activation of AT1R, in particular inflammation, fibrosis and atherosclerosis. The transcription factor nuclear factor κB, a key mediator in inflammation and atherosclerosis, can be activated by angiotensin II through a mechanism that may involve arrestin-dependent AT1 receptor internalization. Peritoneal dialysis is a therapeutic modality for treating patients with end-stage kidney disease. The effectiveness of peritoneal dialysis at removing waste from the circulation is compromised over time as a consequence of peritoneal dialysis-induced peritoneal fibrosis. The non-physiological dialysis solution used in peritoneal dialysis, i.e. highly concentrated, hyperosmotic glucose, acidic pH as well as large volumes infused into the peritoneal cavity, contributes to the development of fibrosis. Numerous trials have been conducted altering certain components of the peritoneal dialysis fluid in hopes of preventing or delaying the fibrotic response with limited success. We hypothesize that structural activation of AT1R by hyperosmotic peritoneal dialysis fluid activates the internalization process and subsequent signaling through the transcription factor nuclear factor κB, resulting in the generation of pro-fibrotic/pro-inflammatory mediators producing peritoneal fibrosis.
Collapse
Affiliation(s)
- Thomas A Morinelli
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, United States.
| | - Louis M Luttrell
- Division of Endocrinology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, United States; Research Service of the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, United States
| | - Erik G Strungs
- Division of Endocrinology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Michael E Ullian
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, United States; Research Service of the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, United States
| |
Collapse
|
29
|
Doura T, Kamiya M, Obata F, Yamaguchi Y, Hiyama TY, Matsuda T, Fukamizu A, Noda M, Miura M, Urano Y. Detection of LacZ-Positive Cells in Living Tissue with Single-Cell Resolution. Angew Chem Int Ed Engl 2016; 55:9620-4. [PMID: 27400827 DOI: 10.1002/anie.201603328] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Indexed: 11/11/2022]
Abstract
The LacZ gene, which encodes Escherichia coli β-galactosidase, is widely used as a marker for cells with targeted gene expression or disruption. However, it has been difficult to detect lacZ-positive cells in living organisms or tissues at single-cell resolution, limiting the utility of existing lacZ reporters. Herein we present a newly developed fluorogenic β-galactosidase substrate suitable for labeling live cells in culture, as well as in living tissues. This precisely functionalized fluorescent probe exhibited dramatic activation of fluorescence upon reaction with the enzyme, remained inside cells by anchoring itself to intracellular proteins, and provided single-cell resolution. Neurons labeled with this probe preserved spontaneous firing, which was enhanced by application of ligands of receptors expressed in the cells, suggesting that this probe would be applicable to investigate functions of targeted cells in living tissues and organisms.
Collapse
Affiliation(s)
- Tomohiro Doura
- Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Mako Kamiya
- Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,PRESTO, Japan, Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan.
| | - Fumiaki Obata
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yoshifumi Yamaguchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,PRESTO, Japan, Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| | - Takeshi Y Hiyama
- Division of Molecular Neurobiology, National Institute for Basic Biology, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan.,School of Life Science, The Graduate University for Advanced Studies, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan
| | - Takashi Matsuda
- School of Life Science, The Graduate University for Advanced Studies, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan
| | - Akiyoshi Fukamizu
- Life Science Center, Tsukuba Advanced Research Alliance, Tsukuba, Ibaraki, 305-8577, Japan.,Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan
| | - Masaharu Noda
- Division of Molecular Neurobiology, National Institute for Basic Biology, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan.,School of Life Science, The Graduate University for Advanced Studies, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi, 444-8787, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,CREST, Japan, Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo, 100-0004, Japan
| | - Yasuteru Urano
- Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,CREST, Japan, Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo, 100-0004, Japan.
| |
Collapse
|
30
|
Doura T, Kamiya M, Obata F, Yamaguchi Y, Hiyama TY, Matsuda T, Fukamizu A, Noda M, Miura M, Urano Y. Detection ofLacZ-Positive Cells in Living Tissue with Single-Cell Resolution. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201603328] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Tomohiro Doura
- Graduate School of Medicine; The University of Tokyo; 7-3-1, Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Mako Kamiya
- Graduate School of Medicine; The University of Tokyo; 7-3-1, Hongo Bunkyo-ku Tokyo 113-0033 Japan
- PRESTO, Japan, Science and Technology Agency; 4-1-8 Honcho Kawaguchi Saitama 332-0012 Japan
| | - Fumiaki Obata
- Graduate School of Pharmaceutical Sciences; The University of Tokyo; 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
| | - Yoshifumi Yamaguchi
- Graduate School of Pharmaceutical Sciences; The University of Tokyo; 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
- PRESTO, Japan, Science and Technology Agency; 4-1-8 Honcho Kawaguchi Saitama 332-0012 Japan
| | - Takeshi Y. Hiyama
- Division of Molecular Neurobiology; National Institute for Basic Biology; 5-1 Higashiyama Myodaiji-cho, Okazaki Aichi 444-8787 Japan
- School of Life Science; The Graduate University for Advanced Studies; 5-1 Higashiyama Myodaiji-cho, Okazaki Aichi 444-8787 Japan
| | - Takashi Matsuda
- School of Life Science; The Graduate University for Advanced Studies; 5-1 Higashiyama Myodaiji-cho, Okazaki Aichi 444-8787 Japan
| | - Akiyoshi Fukamizu
- Life Science Center, Tsukuba Advanced Research Alliance; Tsukuba Ibaraki 305-8577 Japan
- Graduate School of Life and Environmental Sciences; University of Tsukuba; Tsukuba Ibaraki, 305-8577 Japan
| | - Masaharu Noda
- Division of Molecular Neurobiology; National Institute for Basic Biology; 5-1 Higashiyama Myodaiji-cho, Okazaki Aichi 444-8787 Japan
- School of Life Science; The Graduate University for Advanced Studies; 5-1 Higashiyama Myodaiji-cho, Okazaki Aichi 444-8787 Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences; The University of Tokyo; 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
- CREST, Japan, Agency for Medical Research and Development; 1-7-1 Otemachi Chiyoda-ku Tokyo 100-0004 Japan
| | - Yasuteru Urano
- Graduate School of Medicine; The University of Tokyo; 7-3-1, Hongo Bunkyo-ku Tokyo 113-0033 Japan
- Graduate School of Pharmaceutical Sciences; The University of Tokyo; 7-3-1 Hongo Bunkyo-ku Tokyo 113-0033 Japan
- CREST, Japan, Agency for Medical Research and Development; 1-7-1 Otemachi Chiyoda-ku Tokyo 100-0004 Japan
| |
Collapse
|
31
|
Nakano Y, Matoba T, Tokutome M, Funamoto D, Katsuki S, Ikeda G, Nagaoka K, Ishikita A, Nakano K, Koga JI, Sunagawa K, Egashira K. Nanoparticle-Mediated Delivery of Irbesartan Induces Cardioprotection from Myocardial Ischemia-Reperfusion Injury by Antagonizing Monocyte-Mediated Inflammation. Sci Rep 2016; 6:29601. [PMID: 27403534 PMCID: PMC4939605 DOI: 10.1038/srep29601] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/21/2016] [Indexed: 12/19/2022] Open
Abstract
Myocardial ischemia-reperfusion (IR) injury limits the therapeutic effect of early reperfusion therapy for acute myocardial infarction (AMI), in which the recruitment of inflammatory monocytes plays a causative role. Here we develop bioabsorbable poly-lactic/glycolic acid (PLGA) nanoparticles incorporating irbesartan, an angiotensin II type 1 receptor blocker with a peroxisome proliferator-activated receptor (PPAR)γ agonistic effect (irbesartan-NP). In a mouse model of IR injury, intravenous PLGA nanoparticles distribute to the IR myocardium and monocytes in the blood and in the IR heart. Single intravenous treatment at the time of reperfusion with irbesartan-NP (3.0 mg kg(-1) irbesartan), but not with control nanoparticles or irbesartan solution (3.0 mg kg(-1)), inhibits the recruitment of inflammatory monocytes to the IR heart, and reduces the infarct size via PPARγ-dependent anti-inflammatory mechanisms, and ameliorates left ventricular remodeling 21 days after IR. Irbesartan-NP is a novel approach to treat myocardial IR injury in patients with AMI.
Collapse
Affiliation(s)
- Yasuhiro Nakano
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Tetsuya Matoba
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Masaki Tokutome
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Daiki Funamoto
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Shunsuke Katsuki
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Gentaro Ikeda
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kazuhiro Nagaoka
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Ayako Ishikita
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kaku Nakano
- Department of Cardiovascular Research, Development, and Translational Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Jun-Ichiro Koga
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kenji Sunagawa
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kensuke Egashira
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan.,Department of Cardiovascular Research, Development, and Translational Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
32
|
Lu H, Cassis LA, Kooi CWV, Daugherty A. Structure and functions of angiotensinogen. Hypertens Res 2016; 39:492-500. [PMID: 26888118 PMCID: PMC4935807 DOI: 10.1038/hr.2016.17] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 12/13/2022]
Abstract
Angiotensinogen (AGT) is the sole precursor of all angiotensin peptides. Although AGT is generally considered as a passive substrate of the renin-angiotensin system, there is accumulating evidence that the regulation and functions of AGT are intricate. Understanding the diversity of AGT properties has been enhanced by protein structural analysis and animal studies. In addition to whole-body genetic deletion, AGT can be regulated in vivo by cell-specific procedures, adeno-associated viral approaches and antisense oligonucleotides. Indeed, the availability of these multiple manipulations of AGT in vivo has provided new insights into the multifaceted roles of AGT. In this review, the combination of structural and functional studies is highlighted to focus on the increasing recognition that AGT exerts effects beyond being a sole provider of angiotensin peptides.
Collapse
Affiliation(s)
- Hong Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA.,Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Lisa A Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Craig W Vander Kooi
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA.,Department of Physiology, University of Kentucky, Lexington, KY, USA.,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
33
|
Roy A, Dakroub M, Tezini GCSV, Liu Y, Guatimosim S, Feng Q, Salgado HC, Prado VF, Prado MAM, Gros R. Cardiac acetylcholine inhibits ventricular remodeling and dysfunction under pathologic conditions. FASEB J 2015; 30:688-701. [PMID: 26481308 DOI: 10.1096/fj.15-277046] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/28/2015] [Indexed: 12/11/2022]
Abstract
Autonomic dysfunction is a characteristic of cardiac disease and decreased vagal activity is observed in heart failure. Rodent cardiomyocytes produce de novo ACh, which is critical in maintaining cardiac homeostasis. We report that this nonneuronal cholinergic system is also found in human cardiomyocytes, which expressed choline acetyltransferase (ChAT) and the vesicular acetylcholine transporter (VAChT). Furthermore, VAChT expression was increased 3- and 1.5-fold at the mRNA and protein level, respectively, in ventricular tissue from patients with heart failure, suggesting increased ACh secretion in disease. We used mice with genetic deletion of cardiomyocyte-specific VAChT or ChAT and mice overexpressing VAChT to test the functional significance of cholinergic signaling. Mice deficient for VAChT displayed an 8% decrease in fractional shortening and 13% decrease in ejection fraction compared with angiotensin II (Ang II)-treated control animals, suggesting enhanced ventricular dysfunction and pathologic remodeling in response to Ang II. Similar results were observed in ChAT-deficient mice. Conversely, no decline in ventricular function was observed in Ang II-treated VAChT overexpressors. Furthermore, the fibrotic area was significantly greater (P < 0.05) in Ang II-treated VAChT-deficient mice (3.61 ± 0.64%) compared with wild-type animals (2.24 ± 0.11%). In contrast, VAChT overexpressing mice did not display an increase in collagen deposition. Our results provide new insight into cholinergic regulation of cardiac function, suggesting that a compensatory increase in cardiomyocyte VAChT levels may help offset cardiac remodeling in heart failure.
Collapse
Affiliation(s)
- Ashbeel Roy
- *Robarts Research Institute, Department of Physiology and Pharmacology, Department of Medicine, and Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada; Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil; and Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mouhamed Dakroub
- *Robarts Research Institute, Department of Physiology and Pharmacology, Department of Medicine, and Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada; Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil; and Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Geisa C S V Tezini
- *Robarts Research Institute, Department of Physiology and Pharmacology, Department of Medicine, and Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada; Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil; and Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Yin Liu
- *Robarts Research Institute, Department of Physiology and Pharmacology, Department of Medicine, and Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada; Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil; and Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Silvia Guatimosim
- *Robarts Research Institute, Department of Physiology and Pharmacology, Department of Medicine, and Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada; Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil; and Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Qingping Feng
- *Robarts Research Institute, Department of Physiology and Pharmacology, Department of Medicine, and Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada; Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil; and Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Helio C Salgado
- *Robarts Research Institute, Department of Physiology and Pharmacology, Department of Medicine, and Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada; Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil; and Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Vania F Prado
- *Robarts Research Institute, Department of Physiology and Pharmacology, Department of Medicine, and Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada; Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil; and Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Marco A M Prado
- *Robarts Research Institute, Department of Physiology and Pharmacology, Department of Medicine, and Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada; Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil; and Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Robert Gros
- *Robarts Research Institute, Department of Physiology and Pharmacology, Department of Medicine, and Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada; Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil; and Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
34
|
Karnik SS, Unal H, Kemp JR, Tirupula KC, Eguchi S, Vanderheyden PML, Thomas WG. International Union of Basic and Clinical Pharmacology. XCIX. Angiotensin Receptors: Interpreters of Pathophysiological Angiotensinergic Stimuli [corrected]. Pharmacol Rev 2015; 67:754-819. [PMID: 26315714 PMCID: PMC4630565 DOI: 10.1124/pr.114.010454] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The renin angiotensin system (RAS) produced hormone peptides regulate many vital body functions. Dysfunctional signaling by receptors for RAS peptides leads to pathologic states. Nearly half of humanity today would likely benefit from modern drugs targeting these receptors. The receptors for RAS peptides consist of three G-protein-coupled receptors—the angiotensin II type 1 receptor (AT1 receptor), the angiotensin II type 2 receptor (AT2 receptor), the MAS receptor—and a type II trans-membrane zinc protein—the candidate angiotensin IV receptor (AngIV binding site). The prorenin receptor is a relatively new contender for consideration, but is not included here because the role of prorenin receptor as an independent endocrine mediator is presently unclear. The full spectrum of biologic characteristics of these receptors is still evolving, but there is evidence establishing unique roles of each receptor in cardiovascular, hemodynamic, neurologic, renal, and endothelial functions, as well as in cell proliferation, survival, matrix-cell interaction, and inflammation. Therapeutic agents targeted to these receptors are either in active use in clinical intervention of major common diseases or under evaluation for repurposing in many other disorders. Broad-spectrum influence these receptors produce in complex pathophysiological context in our body highlights their role as precise interpreters of distinctive angiotensinergic peptide cues. This review article summarizes findings published in the last 15 years on the structure, pharmacology, signaling, physiology, and disease states related to angiotensin receptors. We also discuss the challenges the pharmacologist presently faces in formally accepting newer members as established angiotensin receptors and emphasize necessary future developments.
Collapse
Affiliation(s)
- Sadashiva S Karnik
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Hamiyet Unal
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Jacqueline R Kemp
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Kalyan C Tirupula
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Satoru Eguchi
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Patrick M L Vanderheyden
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Walter G Thomas
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| |
Collapse
|
35
|
Angiotensin II receptor blockade promotes repair of skeletal muscle through down-regulation of aging-promoting C1q expression. Sci Rep 2015; 5:14453. [PMID: 26571361 PMCID: PMC4585890 DOI: 10.1038/srep14453] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/24/2015] [Indexed: 01/31/2023] Open
Abstract
Disruption of angiotensin II type 1 (AT1) receptor prolonged life span in mice. Since aging-related decline in skeletal muscle function was retarded in Atgr1a−/− mice, we examined the role of AT1 receptor in muscle regeneration after injury. Administration of AT1 receptor blocker irbesartan increased the size of regenerating myofibers, decreased fibrosis, and enhanced functional muscle recovery after cryoinjury. We recently reported that complement C1q, secreted by macrophages, activated Wnt/β-catenin signaling and promoted aging-related decline in regenerative capacity of skeletal muscle. Notably, irbesartan induced M2 polarization of macrophages, but reduced C1q expression in cryoinjured muscles and in cultured macrophage cells. Irbesartan inhibited up-regulation of Axin2, a downstream gene of Wnt/β-catenin pathway, in cryoinjured muscles. In addition, topical administration of C1q reversed beneficial effects of irbesartan on skeletal muscle regeneration after injury. These results suggest that AT1 receptor blockade improves muscle repair and regeneration through down-regulation of the aging-promoting C1q-Wnt/β-catenin signaling pathway.
Collapse
|
36
|
Murata K, Baasanjav A, Kwon C, Hashimoto M, Ishida J, Fukamizu A. Angiotensin II accelerates mammary gland development independently of high blood pressure in pregnancy-associated hypertensive mice. Physiol Rep 2015; 3:3/9/e12542. [PMID: 26341998 PMCID: PMC4600386 DOI: 10.14814/phy2.12542] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Angiotensin II (AngII) is a vasopressor hormone that has critical roles in maintenance of normal blood pressure and pathogenesis of cardiovascular diseases. We previously generated pregnancy-associated hypertensive (PAH) mice by mating female human angiotensinogen transgenic mice with male human renin transgenic mice. PAH mice exhibit hypertension in late pregnancy by overproducing AngII. A recent study demonstrated that angiotensin II type I (AT1) receptor is expressed in mammary epithelial cells and its signaling is critical for mammary gland involution after weaning. However, the role of AngII-AT1 receptor signaling in the development of mammary gland during pregnancy remains unclear. In this study, to investigate the role of AngII-AT1 receptor signaling in mammary gland development during pregnancy, we analyzed the mammary gland of PAH mice. Histological and gene expression analyses revealed that lobuloalveolar development was accelerated with increased milk protein production and lipid accumulation in the mammary gland of PAH mice. Furthermore, AT1 receptor blocker treatment suppressed acceleration of mammary gland development in PAH mice, while the treatment of hydralazine, another antihypertensive drug, did not. These data suggest that AngII-AT1 receptor-induced signaling accelerates mammary gland development during pregnancy through hypertension-independent mechanism.
Collapse
Affiliation(s)
- Kazuya Murata
- Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Altansarnai Baasanjav
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Chulwon Kwon
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Misuzu Hashimoto
- Ph.D. Program in Human Biology, School of Integrative Global Majors (SIGMA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Junji Ishida
- Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akiyoshi Fukamizu
- Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
37
|
|
38
|
Erythropoiesis and Blood Pressure Are Regulated via AT1 Receptor by Distinctive Pathways. PLoS One 2015; 10:e0129484. [PMID: 26107632 PMCID: PMC4479565 DOI: 10.1371/journal.pone.0129484] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 05/08/2015] [Indexed: 11/19/2022] Open
Abstract
The renin–angiotensin system (RAS) plays a central role in blood pressure regulation. Although clinical and experimental studies have suggested that inhibition of RAS is associated with progression of anemia, little evidence is available to support this claim. Here we report that knockout mice that lack angiotensin II, including angiotensinogen and renin knockout mice, exhibit anemia. The anemia of angiotensinogen knockout mice was rescued by angiotensin II infusion, and rescue was completely blocked by simultaneous administration of AT1 receptor blocker. To genetically determine the responsible receptor subtype, we examined AT1a, AT1b, and AT2 knockout mice, but did not observe anemia in any of them. To investigate whether pharmacological AT1 receptor inhibition recapitulates the anemic phenotype, we administered AT1 receptor antagonist in hypotensive AT1a receptor knockout mice to inhibit the remaining AT1b receptor. In these animals, hematocrit levels barely decreased, but blood pressure further decreased to the level observed in angiotensinogen knockout mice. We then generated AT1a and AT1b double-knockout mice to completely ablate the AT1 receptors; the mice finally exhibited the anemic phenotype. These results provide clear evidence that although erythropoiesis and blood pressure are negatively controlled through the AT1 receptor inhibition in vivo, the pathways involved are complex and distinct, because erythropoiesis is more resistant to AT1 receptor inhibition than blood pressure control.
Collapse
|
39
|
Unwrapping the origins and roles of the renal endothelium. Pediatr Nephrol 2015; 30:865-72. [PMID: 24633402 PMCID: PMC4164630 DOI: 10.1007/s00467-014-2798-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 02/18/2014] [Accepted: 02/21/2014] [Indexed: 10/25/2022]
Abstract
The renal vasculature, like all vessels, is lined by a thin layer of simple squamous epithelial cells called an endothelium. These endothelial-lined vessels can be subdivided into four major compartments: arteries, veins, capillaries and lymphatics. The renal vasculature is a highly integrated network that forms through the active processes of angiogenesis and vasculogenesis. Determination of the precise contribution of these two processes and of the molecular signaling that governs the differentiation, specification and maturation of these critical cell populations is the focus of an actively evolving field of research. Although much of the focus has concentrated on the origin of the glomerular capillaries, in this review we extend the investigation to the origins of the endothelial cells throughout the entire kidney and the signaling events that cause their distinct functional and molecular profiles. A thorough understanding of endothelial cell biology may play a critical role in a better understanding of renal vascular diseases.
Collapse
|
40
|
Zempo H, Suzuki JI, Ogawa M, Watanabe R, Isobe M. A different role of angiotensin II type 1a receptor in the development and hypertrophy of plantaris muscle in mice. J Appl Genet 2015; 57:91-7. [DOI: 10.1007/s13353-015-0291-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 04/02/2015] [Accepted: 05/12/2015] [Indexed: 12/25/2022]
|
41
|
Sparks MA, Crowley SD, Gurley SB, Mirotsou M, Coffman TM. Classical Renin-Angiotensin system in kidney physiology. Compr Physiol 2015; 4:1201-28. [PMID: 24944035 DOI: 10.1002/cphy.c130040] [Citation(s) in RCA: 353] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The renin-angiotensin system has powerful effects in control of the blood pressure and sodium homeostasis. These actions are coordinated through integrated actions in the kidney, cardiovascular system and the central nervous system. Along with its impact on blood pressure, the renin-angiotensin system also influences a range of processes from inflammation and immune responses to longevity. Here, we review the actions of the "classical" renin-angiotensin system, whereby the substrate protein angiotensinogen is processed in a two-step reaction by renin and angiotensin converting enzyme, resulting in the sequential generation of angiotensin I and angiotensin II, the major biologically active renin-angiotensin system peptide, which exerts its actions via type 1 and type 2 angiotensin receptors. In recent years, several new enzymes, peptides, and receptors related to the renin-angiotensin system have been identified, manifesting a complexity that was previously unappreciated. While the functions of these alternative pathways will be reviewed elsewhere in this journal, our focus here is on the physiological role of components of the "classical" renin-angiotensin system, with an emphasis on new developments and modern concepts.
Collapse
Affiliation(s)
- Matthew A Sparks
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | | | | | | | | |
Collapse
|
42
|
Ichikawa D, Kamijo-Ikemori A, Sugaya T, Shibagaki Y, Yasuda T, Hoshino S, Katayama K, Igarashi-Migitaka J, Hirata K, Kimura K. Human liver-type fatty acid–binding protein protects against tubulointerstitial injury in aldosterone-induced renal injury. Am J Physiol Renal Physiol 2015; 308:F114-21. [DOI: 10.1152/ajprenal.00469.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
To demonstrate the renoprotective function of human liver-type fatty acid–binding protein (hL-FABP) expressed in proximal tubules in aldosterone (Aldo)-induced renal injury, hL-FABP chromosomal transgenic (Tg) and wild-type (WT) mice received systemic Aldo infusions (Tg-Aldo and WT-Aldo, respectively) were given 1% NaCl water for 28 days. In this model, elevation of systolic blood pressure, monocyte chemoattractant protein-1 expression, macrophage infiltration in the interstitium, tubulointerstitial damage, and depositions of type I and III collagens were observed. Elevation of systolic blood pressure did not differ in WT-Aldo vs. Tg-Aldo animals, however, renal injury was suppressed in Tg-Aldo compared with WT-Aldo mice. Dihydroethidium fluorescence was used to evaluate reactive oxidative stress, which was suppressed in Tg-Aldo compared with WT-Aldo mice. Gene expression of angiotensinogen in the kidney was upregulated, and excretion of urinary angiotensinogen was increased in WT-Aldo mice. This exacerbation was suppressed in Tg-Aldo mice. Expression of hL-FABP was upregulated in proximal tubules of Tg-Aldo mice. Urinary excretion of hL-FABP was significantly greater in Tg-Aldo than in Tg-control mice. In conclusion, hL-FABP ameliorated the tubulointerstitial damage in Aldo-induced renal injury via reducing oxidative stress and suppressing activation of the intrarenal renin-angiotensin system.
Collapse
Affiliation(s)
- Daisuke Ichikawa
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Takashi Yasuda
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Seiko Hoshino
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kimie Katayama
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | | | - Kazuaki Hirata
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kenjiro Kimura
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| |
Collapse
|
43
|
Isegawa K, Hirooka Y, Katsuki M, Kishi T, Sunagawa K. Angiotensin II type 1 receptor expression in astrocytes is upregulated leading to increased mortality in mice with myocardial infarction-induced heart failure. Am J Physiol Heart Circ Physiol 2014; 307:H1448-55. [DOI: 10.1152/ajpheart.00462.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Enhanced central sympathetic outflow worsens left ventricular (LV) remodeling and prognosis in heart failure after myocardial infarction (MI). Previous studies suggested that activation of brain angiotensin II type 1 receptors (AT1R) in the brain stem leads to sympathoexcitation due to neuronal AT1R upregulation. Recent studies, however, revealed the importance of astrocytes for modulating neuronal activity, but whether changes in astrocytes influence central sympathetic outflow in heart failure is unknown. In the normal state, AT1R are only weakly expressed in astrocytes. We hypothesized that AT1R in astrocytes are upregulated in heart failure and modulate the activity of adjacent neurons, leading to enhanced sympathetic outflow. In the present study, by targeting deletion of astrocyte-specific AT1R, we investigated whether AT1R in astrocytes have a key role in enhancing central sympathetic outflow, and thereby influencing LV remodeling process and the prognosis of MI-induced heart failure. Using the Cre-LoxP system, we generated glial fibrillary acidic protein (GFAP)-specific AT1R knockout (GFAP/AT1RKO) mice. Urinary norepinephrine excretion for 24 h, as an indicator of sympathoexcitation, was significantly lower in GFAP/AT1RKO-MI mice than in control-MI mice. LV size and heart weight after MI were significantly smaller in GFAP/AT1RKO mice than in control mice. Prognosis was significantly improved in GFAP/AT1RKO-MI mice compared with control-MI mice. Our findings indicated that AT1R expression was upregulated in brain stem astrocytes in MI-induced heart failure, which worsened LV remodeling and prognosis via sympathoexcitation. Thus, in addition to neuronal AT1R, AT1R in astrocytes appear to have a key role in enhancing central sympathetic outflow in heart failure.
Collapse
Affiliation(s)
- Kengo Isegawa
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yoshitaka Hirooka
- Department of Advanced Cardiovascular Regulation and Therapeutics, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and
| | - Masato Katsuki
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Takuya Kishi
- Department of Advanced Therapeutics for Cardiovascular Diseases, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kenji Sunagawa
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
44
|
Favre GA, Lebrun P, Lopez P, Butori C, Hofman P, Esnault VL, Van Obberghen E. Constitutive activation of the renin-angiotensin system reduces visceral fat and improves glucose tolerance in mice. J Renin Angiotensin Aldosterone Syst 2014; 15:396-409. [PMID: 25371094 DOI: 10.1177/1470320314537695] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION The renin-angiotensin system (RAS), and particularly angiotensin II, is involved in the control of energy balance, glucose homeostasis and kidney functions. The integrated impact of the RAS on glucose homeostasis is still a matter of debate. MATERIALS AND METHODS We used a model of constitutive RAS activation in double transgenic mice (dTGM) carrying both human angiotensinogen and human renin genes. We evaluated energy balance, measured renal functions, performed glucose and insulin tolerance tests, and used ramipril to inhibit the angiotensin-converting enzyme. RESULTS dTGM had a lower physical activity and an increased food intake without change in body weight. Renal impairment was characterized by low-grade albuminuria. High urinary output secondary to polydipsia was associated with proximal tubule dysfunction. Compared to controls, dTGM had a lower hyperglycemia induced by an intraperitoneal glucose administration. This decrease was not due to changes in insulin sensitivity and/or secretion. dTGM had an increased creatinine production and a lower epididymal fat mass. Acute inhibition of angiotensin-converting enzyme with ramipril did not suppress this improved glucose tolerance profile. CONCLUSION Chronic RAS activation is not sufficient to cause insulin resistance in mice. Moreover, adaptation to constitutive RAS activation in mice results in a better glucose tolerance.
Collapse
Affiliation(s)
- Guillaume A Favre
- INSERM, U 1081, Institute for Research on Cancer and Aging of Nice (IRCAN), "Aging and Diabetes" team, France Nephrology Department, University Hospital, Nice, France
| | - Patricia Lebrun
- INSERM, U 1081, Institute for Research on Cancer and Aging of Nice (IRCAN), "Aging and Diabetes" team, France University of Nice-Sophia Antipolis, Nice, France
| | - Pascal Lopez
- INSERM, U 1081, Institute for Research on Cancer and Aging of Nice (IRCAN), "Aging and Diabetes" team, France University of Nice-Sophia Antipolis, Nice, France
| | - Catherine Butori
- Clinical and Experimental Pathology Department, University Hospital, Nice, France
| | - Paul Hofman
- University of Nice-Sophia Antipolis, Nice, France Clinical and Experimental Pathology Department, University Hospital, Nice, France
| | - Vincent Lm Esnault
- INSERM, U 1081, Institute for Research on Cancer and Aging of Nice (IRCAN), "Aging and Diabetes" team, France Nephrology Department, University Hospital, Nice, France
| | - Emmanuel Van Obberghen
- INSERM, U 1081, Institute for Research on Cancer and Aging of Nice (IRCAN), "Aging and Diabetes" team, France University of Nice-Sophia Antipolis, Nice, France Clinical Chemistry Laboratory, University Hospital, Nice, France
| |
Collapse
|
45
|
Yasuno S, Kuwahara K, Kinoshita H, Yamada C, Nakagawa Y, Usami S, Kuwabara Y, Ueshima K, Harada M, Nishikimi T, Nakao K. Angiotensin II type 1a receptor signalling directly contributes to the increased arrhythmogenicity in cardiac hypertrophy. Br J Pharmacol 2014; 170:1384-95. [PMID: 23937445 DOI: 10.1111/bph.12328] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 07/11/2013] [Accepted: 07/21/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Angiotensin II has been implicated in the development of various cardiovascular ailments, including cardiac hypertrophy and heart failure. The fact that inhibiting its signalling reduced the incidences of both sudden cardiac death and heart failure in several large-scale clinical trials suggests that angiotensin II is involved in increased cardiac arrhythmogenicity during the development of heart failure. However, because angiotensin II also promotes structural remodelling, including cardiomyocyte hypertrophy and cardiac fibrosis, it has been difficult to assess its direct contribution to cardiac arrhythmogenicity independently of the structural effects. EXPERIMENTAL APPROACH We induced cardiac hypertrophy in wild-type (WT) and angiotensin II type 1a receptor knockout (AT1aR-KO) mice by transverse aortic constriction (TAC). The susceptibility to ventricular tachycardia (VT) assessed in an in vivo electrophysiological study was compared in the two genotypes. The effect of acute pharmacological blockade of AT1R on the incidences of arrhythmias was also assessed. KEY RESULTS As described previously, WT and AT1aR-KO mice with TAC developed cardiac hypertrophy to the same degree, but the incidence of VT was much lower in the latter. Moreover, although TAC induced an increase in tyrosine phosphorylation of connexin 43, a critical component of gap junctional channels, and a reduction in ventricular levels of connexin 43 protein in both genotypes, the effect was significantly ameliorated in AT1aR-KO mice. Acute pharmacological blockade of AT1R also reduced the incidence of arrhythmias. CONCLUSIONS AND IMPLICATIONS Our findings demonstrate that AT1aR-mediated signalling makes a direct contribution to the increase in arrhythmogenicity in hypertrophied hearts independently of structural remodelling.
Collapse
Affiliation(s)
- Shinji Yasuno
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan; EBM Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Yang X, Zhou H, Qu H, Liu W, Huang X, Shun Y, He L. Effect of Shenxinning decoction on ventricular remodeling in AT1 receptor-knockout mice with chronic renal insufficiency. Indian J Pharmacol 2014; 46:391-7. [PMID: 25097276 PMCID: PMC4118531 DOI: 10.4103/0253-7613.135950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/15/2014] [Accepted: 05/20/2014] [Indexed: 11/06/2022] Open
Abstract
Objective: To observe the efficacy of Shenxinning Decoction (SXND) in ventricular remodeling in AT1 receptor-knockout (AT1-KO) mice with chronic renal insufficiency (CRI). Materials and Methods: AT1-KO mice modeled with subtotal (5/6) nephrectomy were intervened with SXND for 12 weeks. Subsequently, blood urea nitrogen (BUN), serum creatinine (SCr), brain natriuretic peptide (BNP), echocardiography (left ventricular end-diastolic diameter, LVDD; left ventricular end-systolic diameter, LVDS; fractional shortening, FS; and ejection fraction, EF), collagen types I and III in the heart and kidney, myocardial mitochondria, and cardiac transforming growth factor-β1 (TGF-β1) of the AT1-KO mice were compared with the same model with nephrectomy only and untreated with SXND. Results: AT1-KO mice did not affect the process of CRI but it could significantly affect cardiac remodeling process. SXND decreased to some extent the AT1-KO mice's BUN, SCr, BNP, and cardiac LVDD, LVDS, and BNP, improved FS and EF, lowered the expression of collagen type I and III in heart and kidney, increased the quantity of mitochondria and ameliorated their structure, and down-regulated the expression of TGF-β1. Conclusion: SXND may antagonize the renin–angiotensin system (RAS) and decrease uremia toxins, thereby ameliorating ventricular remodeling in CRI. Furthermore, SXND has a mechanism correlated with the improvement of myocardial energy metabolism and the down-regulation of TGF-β1.
Collapse
Affiliation(s)
- Xuejun Yang
- Institute of Kidney Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhou
- Institute of Kidney Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huiyan Qu
- Institute of Kidney Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weifang Liu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojin Huang
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yating Shun
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liqun He
- Institute of Kidney Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
47
|
Schleifenbaum J, Kassmann M, Szijártó IA, Hercule HC, Tano JY, Weinert S, Heidenreich M, Pathan AR, Anistan YM, Alenina N, Rusch NJ, Bader M, Jentsch TJ, Gollasch M. Stretch-activation of angiotensin II type 1a receptors contributes to the myogenic response of mouse mesenteric and renal arteries. Circ Res 2014; 115:263-72. [PMID: 24838176 DOI: 10.1161/circresaha.115.302882] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Vascular wall stretch is the major stimulus for the myogenic response of small arteries to pressure. The molecular mechanisms are elusive, but recent findings suggest that G protein-coupled receptors can elicit a stretch response. OBJECTIVE To determine whether angiotensin II type 1 receptors (AT1R) in vascular smooth muscle cells exert mechanosensitivity and identify the downstream ion channel mediators of myogenic vasoconstriction. METHODS AND RESULTS We used mice deficient in AT1R signaling molecules and putative ion channel targets, namely AT1R, angiotensinogen, transient receptor potential channel 6 (TRPC6) channels, or several subtypes of the voltage-gated K+ (Kv7) gene family (KCNQ3, 4, or 5). We identified a mechanosensing mechanism in isolated mesenteric arteries and in the renal circulation that relies on coupling of the AT1R subtype a to a Gq/11 protein as a critical event to accomplish the myogenic response. Arterial mechanoactivation occurs after pharmacological block of AT1R and in the absence of angiotensinogen or TRPC6 channels. Activation of AT1R subtype a by osmotically induced membrane stretch suppresses an XE991-sensitive Kv channel current in patch-clamped vascular smooth muscle cells, and similar concentrations of XE991 enhance mesenteric and renal myogenic tone. Although XE991-sensitive KCNQ3, 4, and 5 channels are expressed in vascular smooth muscle cells, XE991-sensitive K+ current and myogenic contractions persist in arteries deficient in these channels. CONCLUSIONS Our results provide definitive evidence that myogenic responses of mouse mesenteric and renal arteries rely on ligand-independent, mechanoactivation of AT1R subtype a. The AT1R subtype a signal relies on an ion channel distinct from TRPC6 or KCNQ3, 4, or 5 to enact vascular smooth muscle cell activation and elevated vascular resistance.
Collapse
Affiliation(s)
- Johanna Schleifenbaum
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Mario Kassmann
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - István András Szijártó
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Hantz C Hercule
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Jean-Yves Tano
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Stefanie Weinert
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Matthias Heidenreich
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Asif R Pathan
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Yoland-Marie Anistan
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Natalia Alenina
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Nancy J Rusch
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Michael Bader
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Thomas J Jentsch
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.)
| | - Maik Gollasch
- From the Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC), Berlin, Germany (J.S., M.K., I.A.S., H.C.H., J.-Y.T., Y.-M.A., M.G.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (S.W., M.H., N.A., M.B., T.J.J.); Department Physiology and Pathology of Ion Transport, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (S.W., M.H., T.J.J.); Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock (A.R.P., N.J.R.); and Broad Institute of MIT and Harvard, Cambridge, MA (M.H.).
| |
Collapse
|
48
|
Significance of AT1 receptor independent activation of mineralocorticoid receptor in murine diabetic cardiomyopathy. PLoS One 2014; 9:e93145. [PMID: 24664319 PMCID: PMC3963989 DOI: 10.1371/journal.pone.0093145] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 02/28/2014] [Indexed: 01/19/2023] Open
Abstract
Background Diabetes mellitus (DM) has deleterious influence on cardiac performance independent of coronary artery disease and hypertension. The objective of the present study was to investigate the role of the renin-angiotensin-aldosterone system, especially angiotensin II type 1a receptor (AT1aR) and mineralocorticoid receptor (MR) signaling, in left ventricular (LV) dysfunction induced by diabetes mellitus (DM). Methods and Results DM was induced by intraperitoneal injection of streptozotocin (200 mg/kg BW) in wild-type (WT) or AT1aR knockout (KO) male mice, and they were bred during 6 or 12 weeks. Some KO mice were administered the MR antagonist eplerenone (100 mg/kg body weight). At 6 weeks, LV diastolic function was impaired in WT-DM, but preserved in KO-DM. At that time point MR mRNA expression was upregulated, NADPH oxidase subunit (p47phox) and glutathione peroxidase (GPx1) mRNA expression were upregulated, the staining intensities of LV tissue for 4-hydroxy-2-nonenal was stronger in immunohistochemistry, the number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) positive cells was increased, Bcl-2 protein expression was significantly downregulated, and the expression of SERCA2a and phosphorylated phospholamban was depressed in WT-DM, while these changes were not seen in KO-DM. At 12 weeks, however, these changes were also noted in KO-DM. Eplerenone arrested those changes. The plasma aldosterone concentration was elevated in WT-DM but not in KO-DM at 6 weeks. It showed 3.7-fold elevation at 12 weeks even in KO-DM, which suggests “aldosterone breakthrough” phenomenon. However, the aldosterone content in LV tissue was unchanged in KO-DM. Conclusions DM induced diastolic dysfunction was observed even in KO at 12 weeks, which was ameliorated by minelarocorticoid receptor antagonist, eplerenone. AT1-independent MR activation in the LV might be responsible for the pathogenesis of diabetic cardiomyopathy.
Collapse
|
49
|
Ichikawa D, Kamijo-Ikemori A, Sugaya T, Shibagaki Y, Yasuda T, Katayama K, Hoshino S, Igarashi-Migitaka J, Hirata K, Kimura K. Renoprotective effect of renal liver-type fatty acid binding protein and angiotensin II type 1a receptor loss in renal injury caused by RAS activation. Am J Physiol Renal Physiol 2014; 306:F655-63. [DOI: 10.1152/ajprenal.00460.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The aim of this study was to assess the renoprotective effect of renal human liver-type fatty acid binding protein (hL-FABP) and angiotensin II (ANG II) type 1A receptor (AT1a) loss in renal injury caused by renin-angiotensin system (RAS) activation. We established hL-FABP chromosomal transgenic mice (L-FABP+/−AT1a+/+), crossed the L-FABP+/−AT1a+/+ with AT1a knockdown homo mice (L-FABP−/−AT1a−/−), and generated L-FABP+/−AT1a hetero mice (L-FABP+/−AT1a+/−). After the back-cross of these cubs, L-FABP+/−AT1a−/− were obtained. To activate the renal RAS, wild-type mice (L-FABP−/−AT1a+/+), L-FABP+/−AT1a+/+, L-FABP−/−AT1a+/−, L-FABP+/−AT1a+/−, L-FABP−/−AT1a−/−, and L-FABP+/−AT1a−/− were administered high-dose systemic ANG II infusion plus a high-salt diet for 28 days. In the L-FABP−/−AT1a+/+, RAS activation (L-FABP−/−AT1a+/+RAS) caused hypertension and tubulointerstitial damage. In the L-FABP+/−AT1a+/+RAS, tubulointerstitial damage was significantly attenuated compared with L-FABP−/−AT1a+/+RAS. In the AT1a partial knockout (AT1a+/−) or complete knockout (AT1a−/−) mice, reduction of AT1a expression led to a significantly lower degree of renal injury compared with L-FABP−/−AT1a+/+RAS or L-FABP+/−AT1a+/+RAS mice. Renal injury in L-FABP+/−AT1a+/−RAS mice was significantly attenuated compared with L-FABP−/−AT1a+/−RAS mice. In both L-FABP−/−AT1a−/−RAS and L-FABP+/−AT1a−/−RAS mice, renal damage was rarely found. The degrees of renal hL-FABP expression and urinary hL-FABP levels increased by RAS activation and gradually decreased along with reduction of AT1a expression levels. In conclusion, in this mouse model, renal hL-FABP expression and a decrease in AT1a expression attenuated tubulointerstitial damage due to RAS activation.
Collapse
Affiliation(s)
- Daisuke Ichikawa
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Takashi Yasuda
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Kimie Katayama
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| | - Seiko Hoshino
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | | | - Kazuaki Hirata
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kenjiro Kimura
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan; and
| |
Collapse
|
50
|
Noma K, Toshinai K, Koshinaka K, Nakazato M. Telmisartan suppresses food intake in mice via the melanocortin pathway. Obes Res Clin Pract 2013; 5:e79-e156. [PMID: 24331060 DOI: 10.1016/j.orcp.2010.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 10/29/2010] [Accepted: 11/04/2010] [Indexed: 11/15/2022]
Abstract
SUMMARY Telmisartan, an angiotensin type 1 receptor blocker, is widely used for the treatment of hypertension and related cardiovascular and organ damage. We here describe the effects of telmisartan on food intake and body weight using C57BL/6N mice, KKAy mice that overexpress agouti protein (a mouse model of type 2 diabetes with obesity), and mice deficient for angiotensin II-1a receptor. Telmisartan combined with a high-fat diet significantly reduced food intake and body weight gain in the three groups of mice compared with respective control animals that were fed the high-fat diet without telmisartan. Telmisartan did not induce taste aversion or affect energy expenditure. Intracerebroventricular administration of agouti-related protein, a potent antagonist of the melanocortin 3 receptor (MC3-R) and melanocortin 4 receptor (MC4-R), did not stimulate feeding in telmisartan-treated mice. Telmisartan administration enhanced the alpha-melanocyte stimulating hormone-induced suppression of food intake. This study highlights a potential role for telmisartan in hypothalamic feeding regulation, including melanocortin receptors-mediated suppression of food intake and body weight gain.:
Collapse
Affiliation(s)
| | | | | | - Masamitsu Nakazato
- Division of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| |
Collapse
|