1
|
Aubdool AA, Moyes AJ, Pérez-Ternero C, Baliga RS, Sanghera J, Syed MT, Jaigirdah K, Panesar AK, Tsui JC, Li Y, Vasquez HG, Shen YH, LeMaire SA, Raffort-Lareyre J, Mallat Z, Lu HS, Daugherty A, Hobbs AJ. Endothelium- and Fibroblast-Derived C-Type Natriuretic Peptide Prevents the Development and Progression of Aortic Aneurysms. Arterioscler Thromb Vasc Biol 2025. [PMID: 40177775 DOI: 10.1161/atvbaha.124.322350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Thoracic aortic aneurysm (AA) and abdominal AA are life-threatening diseases characterized by dilation, inflammation, and structural weakness; development of pharmacological therapies is desperately needed. CNP (C-type natriuretic peptide) plays a key role in vascular homeostasis, mediating vasodilator, anti-inflammatory, and antiatherogenic actions. Because such processes drive AA, we determined the role of endogenous CNP in offsetting pathogenesis. METHODS Tissue from patients with AA was analyzed to determine the consequences on CNP signaling. Ascending and suprarenal aortic diameters were assessed at baseline and following Ang II (angiotensin II; 1.44 mg/kg per day) infusion in wild-type, endothelium-restricted (ecCNP-/-), fibroblast-restricted (fbCNP-/-), global CNP (gbCNP-/-), or global NPR-C-/- mice infected with an adeno-associated virus expressing a proprotein convertase subtilisin/kexin type 9 gain-of-function mutation or backcrossed to an apoE-/- background. At 28 days, aortas were harvested for RT-qPCR and histological analyses. CNP (0.2 mg/kg per day) was infused to rescue any adverse phenotype. RESULTS Aneurysmal tissue from patients with thoracic AA and abdominal AA revealed that CNP and NPR-C (natriuretic peptide receptor-C) expression were overtly perturbed. ecCNP-/-, fbCNP-/-, and gbCNP-/- mice exhibited an aggravated phenotype compared with wild-type mice in both ascending and suprarenal aortas, exemplified by greater dilation, fibrosis, elastin degradation, and macrophage infiltration. CNP and NPR-C expression was also dysregulated in murine thoracic AA and abdominal AA, accompanied by increased accumulation of mRNA encoding markers of inflammation, extracellular matrix remodeling/calcification, fibrosis, and apoptosis. CNP also prevented activation of isolated macrophages and vascular smooth muscle cells. An essentially identical phenotype was observed in NPR-C-/- mice and while administration of CNP protected against disease severity in wild-type animals, this phenotypic rescue was not apparent in NPR-C-/- mice. CONCLUSIONS Endothelium- and fibroblast-derived CNP, via NPR-C activation, plays important roles in attenuating AA formation by preserving aortic structure and function. Therapeutic strategies aimed at mimicking CNP bioactivity hold potential to reduce the need for surgical intervention.
Collapse
Affiliation(s)
- Aisah A Aubdool
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Amie J Moyes
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Cristina Pérez-Ternero
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Reshma S Baliga
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Jasspinder Sanghera
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - M Taaha Syed
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Kareemah Jaigirdah
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Anmolpreet K Panesar
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Janice C Tsui
- Division of Surgery and Interventional Science, University College London and Royal Free London NHS Foundation Trust, United Kingdom (J.C.T.)
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.L., H.G.V., Y.H.S., S.A.L.)
| | - Hernan G Vasquez
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.L., H.G.V., Y.H.S., S.A.L.)
| | - Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.L., H.G.V., Y.H.S., S.A.L.)
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.L., H.G.V., Y.H.S., S.A.L.)
- Geisinger Research Institute and Heart & Vascular Institute (S.A.L.)
| | | | - Ziad Mallat
- Division of Cardiovascular Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, United Kingdom (Z.M.)
| | - Hong S Lu
- Department of Physiology, Saha Cardiovascular Research Center, Saha Aortic Center, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Alan Daugherty
- Department of Physiology, Saha Cardiovascular Research Center, Saha Aortic Center, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Adrian J Hobbs
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| |
Collapse
|
2
|
Wang L, Tang Y, Buckley AF, Spurney RF. Podocyte specific knockout of the natriuretic peptide clearance receptor is podocyte protective in focal segmental glomerulosclerosis. PLoS One 2025; 20:e0319424. [PMID: 40063586 PMCID: PMC11892885 DOI: 10.1371/journal.pone.0319424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/31/2025] [Indexed: 05/13/2025] Open
Abstract
Natriuretic peptides (NPs) bind to glomerular podocytes and attenuate glomerular injury. The beneficial effects of NPs are negatively regulated by the NP clearance receptor (NPRC), which is highly expressed in podocytes. To determine if inhibiting NPRC is podocyte protective, we examined the effects of deleting NPRC in both cultured podocytes and in vivo. We found that: 1.Both atrial NP and C-type NP inhibit podocyte apoptosis in cultured podocytes, but these podocyte protective effects are significantly attenuated in cells expressing NPRC, and 2. Atrial NP was significantly more effective than CNP at inhibiting the apoptotic response. Consistent with the protective actions of NPs, podocyte specific knockout of NPRC reduced albuminuria, glomerular sclerosis and tubulointerstitial inflammation in a mouse model of focal segmental glomerulosclerosis. These beneficial actions were associated with: 1. Decreased expression of the myofibroblast marker alpha-smooth muscle actin, 2. Reduced expression of the extracellular matrix proteins collagen 4-alpha-1 and fibronectin, and 3. Preserved expression of the podocyte proteins nephrin and podocin. Inhibiting NP clearance may be a useful therapeutic approach to treat glomerular diseases.
Collapse
MESH Headings
- Podocytes/metabolism
- Podocytes/pathology
- Podocytes/drug effects
- Animals
- Glomerulosclerosis, Focal Segmental/pathology
- Glomerulosclerosis, Focal Segmental/metabolism
- Glomerulosclerosis, Focal Segmental/genetics
- Receptors, Atrial Natriuretic Factor/genetics
- Receptors, Atrial Natriuretic Factor/metabolism
- Receptors, Atrial Natriuretic Factor/deficiency
- Mice
- Apoptosis/drug effects
- Mice, Knockout
- Natriuretic Peptide, C-Type/pharmacology
- Natriuretic Peptide, C-Type/metabolism
- Disease Models, Animal
- Membrane Proteins/metabolism
- Albuminuria/genetics
- Male
- Intracellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Liming Wang
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, United States of America
| | - Yuping Tang
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, United States of America
| | - Anne F. Buckley
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Robert F. Spurney
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, United States of America
| |
Collapse
|
3
|
Sen MG, Chooi R, McMullen JR. Heart-derived factors and organ cross-talk in settings of health and disease: new knowledge and clinical opportunities for multimorbidity. J Physiol 2025. [PMID: 39888058 DOI: 10.1113/jp287400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
Cardiovascular disease affects millions of people worldwide and often presents with other conditions including metabolic, renal and neurological disorders. A variety of secreted factors from multiple organs/tissues (proteins, nucleic acids and lipids) have been implicated in facilitating organ cross-talk that may contribute to the development of multimorbidity. Secreted proteins have received the most attention, with the greatest body of research related to factors released from adipose tissue (adipokines), followed by skeletal muscle (myokines). To date, there have been fewer studies on proteins released from the heart (cardiokines) implicated with organ cross-talk. Early evidence for the secretion of cardiac-specific factors facilitating organ cross-talk came in the form of natriuretic peptides which are secreted via the classical endoplasmic reticulum-Golgi pathway. More recently, studies in cardiomyocyte-specific genetic mouse models have revealed cardiac-initiated organ cross-talk. Cardiomyocyte-specific modulation of microRNAs (miR-208a and miR-23-27-24 cluster) and proteins such as the mediator complex subunit 13 (MED13), G-protein-coupled receptor kinase 2 (GRK2), mutant α-myosin heavy-chain (αMHC), ubiquitin-like modifier-activating enzyme (ATG7), oestrogen receptor alpha (ERα) and fibroblast growth factor 21 (FGF21) have resulted in metabolic and renal phenotypes. These studies have implicated a variety of factors which can be secreted via the classical pathway or via non-classical mechanisms including the release of extracellular vesicles. Cross-talk between the heart and the brain has also been described (e.g. via miR-1 and an emerging concept, interoception: detection of internal neural signals). Here we summarize these studies taking into consideration that factors may be secreted in both settings of health and in disease.
Collapse
Affiliation(s)
- Melodi G Sen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Roger Chooi
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Heart Research Institute, Newtown, New South Wales, Australia
- Monash Alfred Baker Centre for Cardiovascular Research, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
4
|
Lehners M, Schmidt H, Zaldivia MTK, Stehle D, Krämer M, Peter A, Adler J, Lukowski R, Feil S, Feil R. Single-cell analysis identifies the CNP/GC-B/cGMP axis as marker and regulator of modulated VSMCs in atherosclerosis. Nat Commun 2025; 16:429. [PMID: 39814746 PMCID: PMC11735800 DOI: 10.1038/s41467-024-55687-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025] Open
Abstract
A balanced activity of cGMP signaling contributes to the maintenance of cardiovascular homeostasis. Vascular smooth muscle cells (VSMCs) can generate cGMP via three ligand-activated guanylyl cyclases, the NO-sensitive guanylyl cyclase, the atrial natriuretic peptide (ANP)-activated GC-A, and the C-type natriuretic peptide (CNP)-stimulated GC-B. Here, we study natriuretic peptide signaling in murine VSMCs and atherosclerotic lesions. Correlative profiling of pathway activity and VSMC phenotype at the single-cell level shows that phenotypic modulation of contractile VSMCs to chondrocyte-like plaque cells during atherogenesis is associated with a switch from ANP/GC‑A to CNP/GC‑B signaling. Silencing of the CNP/GC-B axis in VSMCs results in an increase of chondrocyte-like plaque cells. These findings indicate that the CNP/GC-B/cGMP pathway is a marker and atheroprotective regulator of modulated VSMCs, limiting their transition to chondrocyte-like cells. Overall, this study highlights the plasticity of cGMP signaling in VSMCs and suggests analogies between CNP-dependent remodeling of bone and blood vessels.
Collapse
MESH Headings
- Animals
- Cyclic GMP/metabolism
- Natriuretic Peptide, C-Type/metabolism
- Natriuretic Peptide, C-Type/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Signal Transduction
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Receptors, Atrial Natriuretic Factor/metabolism
- Receptors, Atrial Natriuretic Factor/genetics
- Single-Cell Analysis
- Male
- Mice, Inbred C57BL
- Biomarkers/metabolism
Collapse
Affiliation(s)
- Moritz Lehners
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Hannes Schmidt
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Maria T K Zaldivia
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Daniel Stehle
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Michael Krämer
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Andreas Peter
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
| | - Julia Adler
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
5
|
Rager C, Klöpper T, Tasch S, Whittaker MR, Exintaris B, Mietens A, Middendorff R. The Influence of Cell Isolation and Culturing on Natriuretic Peptide Receptors in Aortic Vascular Smooth Muscle Cells. Cells 2025; 14:51. [PMID: 39791752 PMCID: PMC11720613 DOI: 10.3390/cells14010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/22/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
Vascular smooth muscle cell (SMC) relaxation by guanylyl cyclases (GCs) and cGMP is mediated by NO and its receptor soluble GC (sGC) or natriuretic peptides (NPs) ANP/BNP and CNP with the receptors GC-A and GC-B, respectively. It is commonly accepted that cultured SMCs differ from those in intact vessels. Nevertheless, cell culture often remains the first step for signaling investigations and drug testing. Previously, we showed that even popular reference genes changed dramatically after SMC isolation from aorta. Regarding NP receptors, a substantial amount of data relies on cell culture. We hypothesize that the NP/cGMP system in intact aortic tunica media differs from isolated and cultured aortic SMCs. Therefore, we studied isolation and culturing effects on the expression of NP receptors GC-A, GC-B, and NP clearance receptor (NPRC) compared to sGC. We investigated intact tunica media and primary SMCs from the longitudinal halves of the same rat aorta. GC activity was monitored by cyclic guanosine monophosphate (cGMP). In addition, we hypothesize that there are sex-dependent differences in the NP/cGMP cascade in both intact tissue and cultured cells. We, therefore, analyzed a male and female cohort. Expression was quantified by RT-qPCR comparing aortic media and SMCs with our recently validated reference gene (RG) small nuclear ribonucleoprotein 2 (U2). Only GC-A was stably expressed. In intact media, GC-A exceeded GC-B and NPRC. However, GC-B, NPRC, and sGC were dramatically upregulated in cultured SMCs of the same aortae different from the stable GC-A. The expression was mirrored by NP-induced GC activity. In cultured cells, changes in GC activity were delayed compared to receptor expression. Minor differences between both sexes could also be revealed. Thus, isolation and culture fundamentally alter the cGMP system in vascular SMCs with potential impact on drug testing and scRNAseq. Especially, the dramatic increase in the clearance receptor NPRC in culture might distort all physiological ANP, BNP, and CNP effects.
Collapse
MESH Headings
- Animals
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Receptors, Atrial Natriuretic Factor/metabolism
- Receptors, Atrial Natriuretic Factor/genetics
- Male
- Rats
- Female
- Aorta/cytology
- Aorta/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/cytology
- Cells, Cultured
- Cyclic GMP/metabolism
- Cell Separation
- Rats, Wistar
- Cell Culture Techniques/methods
Collapse
Affiliation(s)
- Christine Rager
- Institute of Anatomy & Cell Biology, Faculty of Medicine, Justus-Liebig-University, Aulweg 123, 35392 Giessen, Germany; (C.R.)
- Drug Delivery, Disposition, and Dynamics (D4), Monash Institute of Pharmacy & Pharmaceutical Sciences (MIPS), Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia;
| | - Tobias Klöpper
- Institute of Anatomy & Cell Biology, Faculty of Medicine, Justus-Liebig-University, Aulweg 123, 35392 Giessen, Germany; (C.R.)
| | - Sabine Tasch
- Institute of Anatomy & Cell Biology, Faculty of Medicine, Justus-Liebig-University, Aulweg 123, 35392 Giessen, Germany; (C.R.)
| | - Michael Raymond Whittaker
- Drug Delivery, Disposition, and Dynamics (D4), Monash Institute of Pharmacy & Pharmaceutical Sciences (MIPS), Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia;
| | - Betty Exintaris
- Pharmacy and Pharmaceutical Sciences Education, Monash Institute of Pharmacy & Pharmaceutical Sciences (MIPS), Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Andrea Mietens
- Institute of Anatomy & Cell Biology, Faculty of Medicine, Justus-Liebig-University, Aulweg 123, 35392 Giessen, Germany; (C.R.)
| | - Ralf Middendorff
- Institute of Anatomy & Cell Biology, Faculty of Medicine, Justus-Liebig-University, Aulweg 123, 35392 Giessen, Germany; (C.R.)
| |
Collapse
|
6
|
Jiang F, Li X, Lin L, Li M, Zheng J. NPRC promotes hepatic steatosis via USP30-mediated deubiquitination of C/EBPβ. Metabolism 2025; 162:156050. [PMID: 39433172 DOI: 10.1016/j.metabol.2024.156050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/12/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated fatty liver disease (MAFLD) is a prevalent chronic liver condition characterised by dysregulated lipid metabolism. The role of Natriuretic Peptide Receptor C (NPRC), a receptor responsible for clearing natriuretic peptides, in MAFLD remains elusive. Therefore, the aim of the present study was to elucidate the role of NPRC in MAFLD progression. APPROACH AND RESULTS This study demonstrated that NPRC enhanced lipid metabolism reprogramming and accelerated MAFLD progression. Mechanistic investigations, including proteomic and ubiquitination analyses, revealed that elevated NPRC levels stabilized the C/EBPβ protein, leading to excessive lipid accumulation. The DNA-binding domain (DBD) of C/EBPβ interacted with the deubiquitinase USP30, a key regulator that inhibited K149-specific K48-linked polyubiquitination of C/EBPβ. Importantly, the ANPR region of NPRC bound to USP30, facilitating the deubiquitination of C/EBPβ. Furthermore, virtual screening identified punicalin, a natural compound, as a potential inhibitor of NPRC expression, which may reduce hepatic lipid accumulation, inflammation and fibrosis. CONCLUSIONS Our findings indicate that NPRC recruits USP30 to mediate the deubiquitination of C/EBPβ, driving lipid metabolism reprogramming. Targeting NPRC could represent a promising therapeutic approach for MAFLD.
Collapse
Affiliation(s)
- Feng Jiang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Xinmiao Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Lifan Lin
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Mengyuan Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Jianjian Zheng
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| |
Collapse
|
7
|
Perez-Ternero C, Li W, Aubdool AA, Goldin RD, Loy J, Devalia K, Alazawi W, Hobbs AJ. Endogenous C-type natriuretic peptide offsets the pathogenesis of steatohepatitis, hepatic fibrosis, and portal hypertension. PNAS NEXUS 2025; 4:pgae579. [PMID: 39816244 PMCID: PMC11734523 DOI: 10.1093/pnasnexus/pgae579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), hepatic fibrosis, and portal hypertension constitute an increasing public health problem due to the growing prevalence of obesity and diabetes. C-type natriuretic peptide (CNP) is an endogenous regulator of cardiovascular homeostasis, immune cell reactivity, and fibrotic disease. Thus, we investigated a role for CNP in the pathogenesis of MASLD. Wild-type (WT), global CNP (gbCNP-/-), and natriuretic peptide receptor-C (NPR-C-/-) knockout mice were fed a choline-deficient defined amino acid diet or administered CCl4. Liver damage was assessed by histological and biochemical analyses, with steatosis and portal vein size determined by ultrasound. Portal vein pressure and reactivity were measured in vivo and ex vivo, respectively. Pharmacological CNP delivery was used to evaluate prospective therapeutic benefit, and plasma CNP concentration was compared in controls and patients with cirrhosis. Circulating CNP concentration was lower in patients with cirrhosis compared with controls. gbCNP-/- mice were more susceptible, versus WT, to advanced steatohepatitis and hepatic fibrosis, characterized by increased immune cell infiltration, fibrosis, ballooning, plasma alanine aminotransferase concentration, and up-regulation of markers driving these processes. gbCNP-/- mice had increased portal vein diameter and pressure, underpinned by CNP insensitivity. NPR-C-/- animals recapitulated, comparatively, the exaggerated pathogenic phenotype in gbCNP-/- mice, whereas CNP reduced hepatic stellate cell proliferation via NPR-B-dependent inhibition of extracellular signal-related kinase 1/2. Administration of CNP reversed many aspects of disease severity. These data define a new intrinsic role for CNP in offsetting the pathogenesis of MASLD, hepatic fibrosis, and portal hypertension and the potential for targeting CNP signaling for treating these disorders.
Collapse
Affiliation(s)
- Cristina Perez-Ternero
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Wenhao Li
- Barts Liver Centre, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Aisah A Aubdool
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Robert D Goldin
- Centre for Pathology, St Mary’s Hospital, Imperial College, London W2 1NY, United Kingdom
| | - John Loy
- Bariatric Surgery Department, Homerton University Hospital, Homerton Row, London E9 6SR, United Kingdom
| | - Kalpana Devalia
- Bariatric Surgery Department, Homerton University Hospital, Homerton Row, London E9 6SR, United Kingdom
| | - William Alazawi
- Barts Liver Centre, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Adrian J Hobbs
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| |
Collapse
|
8
|
Li XC, Wang CH, Hassan R, Katsurada A, Sato R, Zhuo JL. Deletion of AT 1a receptors selectively in the proximal tubules of the kidney alters the hypotensive and natriuretic response to atrial natriuretic peptide via NPR A/cGMP/NO signaling. Am J Physiol Renal Physiol 2024; 327:F946-F956. [PMID: 39361722 PMCID: PMC11687850 DOI: 10.1152/ajprenal.00160.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
In the proximal tubules of the kidney, angiotensin II (ANG II) binds and activates ANG II type 1 (AT1a) receptors to stimulate proximal tubule Na+ reabsorption, whereas atrial natriuretic peptide (ANP) binds and activates natriuretic peptide receptors (NPRA) to inhibit ANG II-induced proximal tubule Na+ reabsorption. These two vasoactive systems play important counteracting roles to control Na+ reabsorption in the proximal tubules and help maintain blood pressure homeostasis. However, how AT1a and NPRA receptors interact in the proximal tubules and whether natriuretic effects of NPRA receptor activation by ANP may be potentiated by deletion of AT1 (AT1a) receptors selectively in the proximal tubules have not been studied previously. The present study used a novel mouse model with proximal tubule-specific knockout of AT1a receptors, PT-Agtr1a-/-, to test the hypothesis that deletion of AT1a receptors selectively in the proximal tubules augments the hypotensive and natriuretic responses to ANP. Basal blood pressure was about 16 ± 3 mmHg lower (P < 0.01), fractional proximal tubule Na+ reabsorption was significantly lower (P < 0.05), whereas 24-h urinary Na+ excretion was significantly higher, in PT-Agtr1a-/- mice than in wild-type mice (P < 0.01). Infusion of ANP via osmotic minipump for 2 wk (0.5 mg/kg/day ip) further significantly decreased blood pressure and increased the natriuretic response in PT-Agtr1a-/- mice by inhibiting proximal tubule Na+ reabsorption compared with wild-type mice (P < 0.01). These augmented hypotensive and natriuretic responses to ANP in PT-Agtr1a-/- mice were associated with increased plasma and kidney cGMP levels (P < 0.01), kidney cortical NPRA and NPRC mRNA expression (P < 0.05), endothelial nitric oxide (NO) synthase (eNOS) and phosphorylated eNOS proteins (P < 0.01), and urinary NO excretion (P < 0.01). Taken together, the results of the present study provide further evidence for important physiological roles of intratubular ANG II/AT1a and ANP/NPRA signaling pathways in the proximal tubules to regulate proximal tubule Na+ reabsorption and maintain blood pressure homeostasis.NEW & NOTEWORTHY This study used a mutant mouse model with proximal tubule-selective deletion of angiotensin II (ANG II) type 1 (AT1a) receptors to study, for the first time, important interactions between ANG II/AT1 (AT1a) receptor/Na+/H+ exchanger 3 and atrial natriuretic peptide (ANP)/natriuretic peptide receptor (NPRA)/cGMP/nitric oxide signaling pathways in the proximal tubules. The results of the present study provide further evidence for important physiological roles of proximal tubule ANG II/AT1a and ANP/NPRA signaling pathways in the regulation of proximal tubule Na+ reabsorption and blood pressure homeostasis.
Collapse
MESH Headings
- Animals
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/drug effects
- Cyclic GMP/metabolism
- Atrial Natriuretic Factor/metabolism
- Atrial Natriuretic Factor/genetics
- Receptors, Atrial Natriuretic Factor/metabolism
- Receptors, Atrial Natriuretic Factor/genetics
- Receptors, Atrial Natriuretic Factor/deficiency
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Mice, Knockout
- Signal Transduction
- Nitric Oxide/metabolism
- Blood Pressure/drug effects
- Male
- Natriuresis/drug effects
- Sodium/metabolism
- Sodium/urine
- Hypotension/metabolism
- Hypotension/genetics
- Hypotension/physiopathology
- Renal Reabsorption/drug effects
- Mice
- Nitric Oxide Synthase Type III/metabolism
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Xiao Chun Li
- Tulane Hypertension and Renal Center of Excellence and Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Chih-Hong Wang
- Tulane Hypertension and Renal Center of Excellence and Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Rumana Hassan
- Tulane Hypertension and Renal Center of Excellence and Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Akemi Katsurada
- Tulane Hypertension and Renal Center of Excellence and Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Ryosuke Sato
- Tulane Hypertension and Renal Center of Excellence and Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Jia Long Zhuo
- Tulane Hypertension and Renal Center of Excellence and Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States
| |
Collapse
|
9
|
Nakamori T, Komatsuzawa I, Iwata U, Makita A, Kagiya G, Fujitani K, Kitaguchi T, Tsuboi T, Ohki-Hamazaki H. The role of osteocrin in memory formation during early learning, as revealed by visual imprinting in chicks. iScience 2024; 27:111195. [PMID: 39600306 PMCID: PMC11591550 DOI: 10.1016/j.isci.2024.111195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/17/2024] [Accepted: 10/15/2024] [Indexed: 11/29/2024] Open
Abstract
Osteocrin (OSTN) is structurally associated with natriuretic peptides. Its expression in the brain, which has only been recognized in anthropoid primates, is induced by sensory stimuli and regulates the activity-dependent dendritic growth of neurons. However, details on the signaling mechanisms of OSTN and its function in plastic changes during learning and memory have yet to be elucidated. We found that OSTN was expressed in the cortical region of the chicken brain. The injection of chicken OSTN (chOSTN) after imprinting training prolonged the memory retention for the imprinting stimulus. Conversely, a reduction in the OSTN receptor chNPR3 inhibited memory retention. The memory retention was positively correlated with a high level of chOSTN and fewer neurites in the cortical region. In conclusion, OSTN-NPR3 signaling promoted memory consolidation and/or retention by regulating neurite branching during childhood.
Collapse
Affiliation(s)
- Tomoharu Nakamori
- College of Liberal Arts and Sciences, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Izumi Komatsuzawa
- College of Liberal Arts and Sciences, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Umi Iwata
- College of Liberal Arts and Sciences, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Ami Makita
- College of Liberal Arts and Sciences, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Go Kagiya
- School of Allied Health Sciences, and Regenerative Medicine and Cell Design Research Facility, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - Kazuko Fujitani
- Gene Analysis Center, School of Medicine, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - Tetsuya Kitaguchi
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Hiroko Ohki-Hamazaki
- College of Liberal Arts and Sciences, Kitasato University, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| |
Collapse
|
10
|
Jensen M, Heinl ES, Federlein A, Schwartz U, Lund L, Madsen K, Jensen BL, Schweda F. Identification of natriuretic peptide receptor A-related gene expression signatures in podocytes in vivo reveals baseline control of protective pathways. Am J Physiol Renal Physiol 2024; 327:F806-F821. [PMID: 39298549 DOI: 10.1152/ajprenal.00394.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024] Open
Abstract
Natriuretic peptide receptor-A (NPR-A) is the principal receptor for the natriuretic peptides atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). Targeted deletion of NPR-A in mouse glomerular podocytes significantly enhances renal injury in vivo in the DOCA-salt experimental model. It was therefore hypothesized that natriuretic peptides exert a direct protective effect on glomerular barrier integrity through activation of NPR-A and modulation of gene expression patterns in podocytes. Green fluorescence-positive podocytes from mice with a conditional deletion of Npr1 encoding NPR-A were isolated by fluorescence-activated cell sorting (FACS). Differentially expressed genes (DEGs) in podocytes were identified by RNA sequencing of podocytes from wild-type and NPR-A-deleted mice. Enrichment analysis was performed on the DEGs using Gene Ontology (GO) terms. Identified transcripts were validated by real-time PCR and ELISA of cultured isolated human and mouse glomeruli. In addition, the effect of natriuretic peptides on podocyte migration was investigated by measuring the outgrowth of podocytes from cultured glomeruli. A total of 158 DEGs were identified with 81 downregulated DEGs and 77 upregulated DEGs in Npr1-deficient podocytes. Among the downregulated genes were protein S and semaphorin 3G, which are known to have protective effects in podocytes. Protein S was also expressed in and secreted from isolated human glomeruli. GO enrichment analysis revealed that the upregulated DEGs in NPR-A deficient podocytes were associated with cell migration and motility. In line, BNP significantly decreased podocyte outgrowth from cultured glomeruli. In conclusion, endogenous levels of natriuretic peptides in mice support baseline protective pathways at glomerular podocytes such as protein S and suppress podocyte migration.NEW & NOTEWORTHY A combination of fluorescence-activated cell sorting and RNA sequencing identified 158 changed gene products in adult mouse kidneys with and without podocyte-specific deletion of the natriuretic peptide receptor A. Downregulated products included protein S and semaphorin 3G, both with proven renoprotective impact, whereas upregulated products were related to mobility of podocytes. Protein S was produced and released from human and murine isolated glomeruli, and atrial natriuretic peptide (ANP) led to decreased migration of podocytes.
Collapse
Affiliation(s)
- Mia Jensen
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Elena-Sofia Heinl
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Anna Federlein
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Uwe Schwartz
- NGS Analysis Center, Biology and Pre-Clinical Medicine, University of Regensburg, Regensburg, Germany
| | - Lars Lund
- Department of Urology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Kirsten Madsen
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Boye L Jensen
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Frank Schweda
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
11
|
Lowe VJ, Aubdool AA, Moyes AJ, Dignam JP, Perez-Ternero C, Baliga RS, Smart N, Hobbs AJ. Cardiomyocyte-derived C-type natriuretic peptide diminishes myocardial ischaemic injury by promoting revascularisation and limiting fibrotic burden. Pharmacol Res 2024; 209:107447. [PMID: 39374886 DOI: 10.1016/j.phrs.2024.107447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND C-type natriuretic peptide (CNP) is a significant player in the maintenance of cardiac and vascular homeostasis regulating local blood flow, platelet and leukocyte activation, heart structure and function, angiogenesis and metabolic balance. Since such processes are perturbed in myocardial infarction (MI), we explored the role of cardiomyocyte-derived CNP, and pharmacological administration of the peptide, in offsetting the pathological consequences of MI. METHODS Wild type (WT) and cardiomyocyte-restricted CNP null (cmCNP-/-) mice were subjected to left anterior descending coronary artery (LADCA) ligation and acute effects on infarct size and longer-term outcomes of cardiac repair explored. Heart structure and function were assessed by combined echocardiographic and molecular analyses. Pharmacological administration of CNP (0.2 mg/kg/day; s.c.) was utilized to assess therapeutic potential. RESULTS Compared to WT littermates, cmCNP-/- mice had a modestly increased infarct size following LADCA ligation but without significant deterioration of cardiac structural and functional indices. However, cmCNP-/- animals exhibited overtly worse heart morphology and contractility 6 weeks following MI, with particularly deleterious reductions in left ventricular ejection fraction, dilatation, fibrosis and revascularization. This phenotype was largely recapitulated in animals with global deletion of natriuretic peptide receptor (NPR)-C (NPR-C-/-). Pharmacological administration of CNP rescued the deleterious pathology in WT and cmCNP-/-, but not NPR-C-/-, animals. CONCLUSIONS AND IMPLICATIONS Cardiomyocytes synthesize and release CNP as an intrinsic protective mechanism in response to MI that reduces cardiac structural and functional deficits; these salutary actions are primarily NPR-C-dependent. Pharmacological targeting of CNP may represent a new therapeutic option for MI.
Collapse
Affiliation(s)
- Vanessa J Lowe
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Aisah A Aubdool
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Amie J Moyes
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Joshua P Dignam
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - C Perez-Ternero
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Reshma S Baliga
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Nicola Smart
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX3 7TY, UK
| | - Adrian J Hobbs
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
12
|
Weber CJ, Weitzel AJ, Liu AY, Gacasan EG, Sah RL, Cooper KL. Cellular and molecular mechanisms that shape the development and evolution of tail vertebral proportion in mice and jerboas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620311. [PMID: 39484405 PMCID: PMC11527341 DOI: 10.1101/2024.10.25.620311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Despite the functional importance of the vertebral skeleton, little is known about how individual vertebrae elongate or achieve disproportionate lengths as in the giraffe neck. Rodent tails are an abundantly diverse and more tractable system to understand mechanisms of vertebral growth and proportion. In many rodents, disproportionately long mid-tail vertebrae form a 'crescendo-decrescendo' of lengths in the tail series. In bipedal jerboas, these vertebrae grow exceptionally long such that the adult tail is 1.5x the length of a mouse tail, relative to body length, with four fewer vertebrae. How do vertebrae with the same regional identity elongate differently from their neighbors to establish and diversify adult proportion? Here, we find that vertebral lengths are largely determined by differences in growth cartilage height and the number of cells progressing through endochondral ossification. Hypertrophic chondrocyte size, a major contributor to differential elongation in mammal limb bones, differs only in the longest jerboa mid-tail vertebrae where they are exceptionally large. To uncover candidate molecular mechanisms of disproportionate vertebral growth, we performed intersectional RNA-Seq of mouse and jerboa tail vertebrae with similar and disproportionate elongation rates. Many regulators of posterior axial identity and endochondral elongation are disproportionately differentially expressed in jerboa vertebrae. Among these, the inhibitory natriuretic peptide receptor C (NPR3) appears in multiple studies of rodent and human skeletal proportion suggesting it refines local growth rates broadly in the skeleton and broadly in mammals. Consistent with this hypothesis, NPR3 loss of function mice have abnormal tail and limb proportions. Therefore, in addition to genetic components of the complex process of vertebral evolution, these studies reveal fundamental mechanisms of skeletal growth and proportion.
Collapse
Affiliation(s)
- Ceri J Weber
- Department of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Alexander J Weitzel
- Department of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Alexander Y Liu
- Department of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Erica G Gacasan
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Robert L Sah
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Kimberly L Cooper
- Department of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
13
|
Carper D, Lac M, Coue M, Labour A, Märtens A, Banda JAA, Mazeyrie L, Mechta M, Ingerslev LR, Elhadad M, Petit JV, Maslo C, Monbrun L, Del Carmine P, Sainte-Marie Y, Bourlier V, Laurens C, Mithieux G, Joanisse DR, Coudray C, Feillet-Coudray C, Montastier E, Viguerie N, Tavernier G, Waldenberger M, Peters A, Wang-Sattler R, Adamski J, Suhre K, Gieger C, Kastenmüller G, Illig T, Lichtinghagen R, Seissler J, Mounier R, Hiller K, Jordan J, Barrès R, Kuhn M, Pesta D, Moro C. Loss of atrial natriuretic peptide signaling causes insulin resistance, mitochondrial dysfunction, and low endurance capacity. SCIENCE ADVANCES 2024; 10:eadl4374. [PMID: 39383215 PMCID: PMC11463261 DOI: 10.1126/sciadv.adl4374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 09/06/2024] [Indexed: 10/11/2024]
Abstract
Type 2 diabetes (T2D) and obesity are strongly associated with low natriuretic peptide (NP) plasma levels and a down-regulation of NP guanylyl cyclase receptor-A (GCA) in skeletal muscle and adipose tissue. However, no study has so far provided evidence for a causal link between atrial NP (ANP)/GCA deficiency and T2D pathogenesis. Here, we show that both systemic and skeletal muscle ANP/GCA deficiencies in mice promote metabolic disturbances and prediabetes. Skeletal muscle insulin resistance is further associated with altered mitochondrial function and impaired endurance running capacity. ANP/GCA-deficient mice exhibit increased proton leak and reduced content of mitochondrial oxidative phosphorylation proteins. We further show that GCA is related to several metabolic traits in T2D and positively correlates with markers of oxidative capacity in human skeletal muscle. Together, these results indicate that ANP/GCA signaling controls muscle mitochondrial integrity and oxidative capacity in vivo and plays a causal role in the development of prediabetes.
Collapse
Affiliation(s)
- Deborah Carper
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Marlène Lac
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Marine Coue
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Axel Labour
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Andre Märtens
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig and Physikalisch-Technische Bundesanstalt, Brunswick, Germany
| | - Jorge Alberto Ayala Banda
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Laurène Mazeyrie
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Mie Mechta
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Roed Ingerslev
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mohamed Elhadad
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | | | - Claire Maslo
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Laurent Monbrun
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Peggy Del Carmine
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1315, CNRS UMR, 5261 Lyon, France
| | - Yannis Sainte-Marie
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Virginie Bourlier
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Claire Laurens
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | | | - Denis R. Joanisse
- Department of Kinesiology, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Canada
| | - Charles Coudray
- Dynamique Musculaire Et Métabolisme, INRAE, UMR866, Université Montpellier, Montpellier, France
| | | | - Emilie Montastier
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Nathalie Viguerie
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Geneviève Tavernier
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| | - Melanie Waldenberger
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Rui Wang-Sattler
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, Hanover, Germany
| | - Ralf Lichtinghagen
- Department of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Jochen Seissler
- Diabetes Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU, München, Germany
| | - Remy Mounier
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1315, CNRS UMR, 5261 Lyon, France
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig and Physikalisch-Technische Bundesanstalt, Brunswick, Germany
| | - Jens Jordan
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
- Medical Faculty, University of Cologne, Cologne, Germany
| | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Dominik Pesta
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
- Medical Faculty, University of Cologne, Cologne, Germany
- Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Cedric Moro
- Institute of Metabolic and Cardiovascular Diseases, INSERM/Paul Sabatier University, UMR1297, Team MetaDiab, Toulouse, France
| |
Collapse
|
14
|
Dickinson YA, Moyes AJ, Hobbs AJ. C-type natriuretic peptide (CNP): The cardiovascular system and beyond. Pharmacol Ther 2024; 262:108708. [PMID: 39154787 DOI: 10.1016/j.pharmthera.2024.108708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
C-type natriuretic peptide (CNP) represents the 'local' member of the natriuretic peptide family, functioning in an autocrine or paracrine capacity to modulate a hugely diverse portfolio of physiological processes. Whilst the best-characterised of these regulatory roles are in the cardiovascular system, akin to its predominantly endocrine siblings atrial (ANP) and brain (BNP) natriuretic peptides, CNP governs many additional, unrelated mechanisms including bone growth, gamete maturation, auditory processing, and neuronal integrity. Furthermore, there is currently great interest in mimicking the biological activity of CNP for therapeutic gain in many of these disparate organ systems. Herein, we provide an overview of the physiology, pathophysiology and pharmacology of CNP in both cardiovascular and non-cardiovascular settings.
Collapse
Affiliation(s)
- Yasmin A Dickinson
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Amie J Moyes
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Adrian J Hobbs
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
15
|
Gruson D, Hammerer-Lercher A, Collinson P, Duff C, Baum H, Pulkki K, Suvisaari J, Stankovic S, Laitinen P, Bayes-Genis A. The multidimensional value of natriuretic peptides in heart failure, integrating laboratory and clinical aspects. Crit Rev Clin Lab Sci 2024; 61:458-472. [PMID: 38523480 DOI: 10.1080/10408363.2024.2319578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/15/2024] [Accepted: 02/13/2024] [Indexed: 03/26/2024]
Abstract
Natriuretic peptides (NP) play an essential role in heart failure (HF) regulation, and their measurement has improved diagnostic and prognostic accuracy. Clinical symptoms and objective measurements, such as NP levels, should be included in the HF definition to render it more reliable and consistent among observers, hospitals, and healthcare systems. BNP and NT-proBNP are reasonable surrogates for cardiac disease, and their measurement is critical to early diagnosis and risk stratification of HF patients. NPs should be measured in all patients presenting with dyspnea or other symptoms suggestive of HF to facilitate early diagnosis and risk stratification. Both BNP and NT-proBNP are currently used for guided HF management and display comparable diagnostic and prognostic accuracy. Standardized cutoffs for each NP assay are essential for data comparison. The value of NP testing is recognized at various levels, including patient empowerment and education, analytical and operational issues, clinical HF management, and cost-effectiveness.
Collapse
Affiliation(s)
- Damien Gruson
- Pôle de recherche en Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Cliniques Universitaires St-Luc and Université Catholique de Louvain, Brussels, Belgium
- Department of Clinical Biochemistry, Cliniques Universitaires St-Luc and Université Catholique de Louvain, Brussels, Belgium
| | | | - Paul Collinson
- Department of Clinical Blood Science Chemical Pathology and Cardiology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Christopher Duff
- Department of Clinical Biochemistry, University Hospitals of North Midlands, Stoke-on-Trent, UK
| | - Hannsjörg Baum
- Department Laboratory Medicine, Regionale Kliniken Holding RKH, Ludwigsburg, Germany
| | - Kari Pulkki
- Clinical Chemistry and Hematology, Diagnostic Center, Helsinki University Hospital and the University of Helsinki, Helsinki, Finland
| | - Janne Suvisaari
- Clinical Chemistry and Hematology, Diagnostic Center, Helsinki University Hospital and the University of Helsinki, Helsinki, Finland
| | - Sanja Stankovic
- Center for Medical Biochemistry, University Clinical Center of Serbia, Belgrade, Serbia
- Department of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Paivi Laitinen
- Center for Medical Biochemistry, University Clinical Center of Serbia, Belgrade, Serbia
| | - Antoni Bayes-Genis
- Germans Trias Heart Institute (iCor), Pujol, Universitat Autònoma de Barcelona; CIBERCV, Barcelona, Spain
| |
Collapse
|
16
|
Bie P. Plasma concentrations of peptide hormones: Unrealistic levels of vasopressin (AVP), oxytocin (OXT), and brain natriuretic peptide (BNP). Acta Physiol (Oxf) 2024; 240:e14200. [PMID: 39034759 DOI: 10.1111/apha.14200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024]
Abstract
Hormones are specific molecules measured in biological fluids by elaborate analytical systems requiring meticulous attention. Variation between laboratories can be expected. However, recently published measurements of AVP, OXT, and BNP in human plasma under basal/control conditions include numbers which, between publications, vary by 100-10 000-fold. Generally, the methods descriptions are scant, at best, and provide no information about quality control measures. Clearly, two results describing the same basal hormone concentration by numbers three orders of magnitude apart are incongruent providing reason for concern. Basal concentrations of bioactive AVP, OXT, and BNP in human plasma are in the order of 1-10 pmol/L. Therefore, assay systems applied to plasma must be able to measure concentrations of less than 1 pmol/L with appropriate specificity and accuracy. Basal concentrations of AVP, OXT, and BNP above 100 pmol/L should be reconsidered, as such results do not reflect bioactive hormone levels in humans, rats, or mice. Any concentration above 1000 pmol/L is of concern because such levels of bioactive hormone may be seen only under extreme conditions, if at all.
Collapse
Affiliation(s)
- Peter Bie
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
17
|
Li SJ, Wei JQ, Kang YY, Wang RQ, Rong WW, Zhao JJ, Deng QW, Gao PJ, Li XD, Wang JG. Natriuretic peptide receptor-C perturbs mitochondrial respiration in white adipose tissue. J Lipid Res 2024; 65:100623. [PMID: 39154732 PMCID: PMC11418126 DOI: 10.1016/j.jlr.2024.100623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/15/2024] [Accepted: 08/11/2024] [Indexed: 08/20/2024] Open
Abstract
Natriuretic peptide receptor-C (NPR-C) is highly expressed in adipose tissues and regulates obesity-related diseases; however, the detailed mechanism remains unknown. In this research, we aimed to explore the potential role of NPR-C in cold exposure and high-fat/high-sugar (HF/HS) diet-induced metabolic changes, especially in regulating white adipose tissue (WAT) mitochondrial function. Our findings showed that NPR-C expression, especially in epididymal WAT (eWAT), was reduced after cold exposure. Global Npr3 (gene encoding NPR-C protein) deficiency led to reduced body weight, increased WAT browning, thermogenesis, and enhanced expression of genes related to mitochondrial biogenesis. RNA-sequencing of eWAT showed that Npr3 deficiency enhanced the expression of mitochondrial respiratory chain complex genes and promoted mitochondrial oxidative phosphorylation in response to cold exposure. In addition, Npr3 KO mice were able to resist obesity induced by HF/HS diet. Npr3 knockdown in stromal vascular fraction (SVF)-induced white adipocytes promoted the expression of proliferator-activated receptor gamma coactivator 1α (PGC1α), uncoupling protein one (UCP1), and mitochondrial respiratory chain complexes. Mechanistically, NPR-C inhibited cGMP and calcium signaling in an NPR-B-dependent manner but suppressed cAMP signaling in an NPR-B-independent manner. Moreover, Npr3 knockdown induced browning via AKT and p38 pathway activation, which were attenuated by Npr2 knockdown. Importantly, treatment with the NPR-C-specific antagonist, AP-811, decreased WAT mass and increased PGC-1α, UCP1, and mitochondrial complex expression. Our findings reveal that NPR-C deficiency enhances mitochondrial function and energy expenditure in white adipose tissue, contributing to improved metabolic health and resistance to obesity.
Collapse
Affiliation(s)
- Shi-Jin Li
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Jin-Qiu Wei
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan-Yuan Kang
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui-Qi Wang
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wu-Wei Rong
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia-Jia Zhao
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian-Wan Deng
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Dong Li
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ji-Guang Wang
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Werner F, Naruke T, Sülzenbrück L, Schäfer S, Rösch M, Völker K, Krebes L, Abeßer M, Möllmann D, Baba HA, Schweda F, Zernecke A, Kuhn M. Auto/Paracrine C-Type Natriuretic Peptide/Cyclic GMP Signaling Prevents Endothelial Dysfunction. Int J Mol Sci 2024; 25:7800. [PMID: 39063044 PMCID: PMC11277478 DOI: 10.3390/ijms25147800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Endothelial dysfunction is cause and consequence of cardiovascular diseases. The endothelial hormone C-type natriuretic peptide (CNP) regulates vascular tone and the vascular barrier. Its cGMP-synthesizing guanylyl cyclase-B (GC-B) receptor is expressed in endothelial cells themselves. To characterize the role of endothelial CNP/cGMP signaling, we studied mice with endothelial-selective GC-B deletion. Endothelial EC GC-B KO mice had thicker, stiffer aortae and isolated systolic hypertension. This was associated with increased proinflammatory E-selectin and VCAM-1 expression and impaired nitric oxide bioavailability. Atherosclerosis susceptibility was evaluated in such KO and control littermates on Ldlr (low-density lipoprotein receptor)-deficient background fed a Western diet for 10 weeks. Notably, the plaque areas and heights within the aortic roots were markedly increased in the double EC GC-B/Ldlr KO mice. This was accompanied by enhanced macrophage infiltration and greater necrotic cores, indicating unstable plaques. Finally, we found that EC GC-B KO mice had diminished vascular regeneration after critical hind-limb ischemia. Remarkably, all these genotype-dependent changes were only observed in female and not in male mice. Auto/paracrine endothelial CNP/GC-B/cGMP signaling protects from arterial stiffness, systolic hypertension, and atherosclerosis and improves reparative angiogenesis. Interestingly, our data indicate a sex disparity in the connection of diminished CNP/GC-B activity to endothelial dysfunction.
Collapse
MESH Headings
- Animals
- Natriuretic Peptide, C-Type/metabolism
- Natriuretic Peptide, C-Type/genetics
- Cyclic GMP/metabolism
- Mice
- Male
- Mice, Knockout
- Signal Transduction
- Female
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Receptors, Atrial Natriuretic Factor/metabolism
- Receptors, Atrial Natriuretic Factor/genetics
- Endothelial Cells/metabolism
- Receptors, LDL/metabolism
- Receptors, LDL/genetics
- Paracrine Communication
- Hypertension/metabolism
- Hypertension/genetics
- Mice, Inbred C57BL
- Aorta/metabolism
- Aorta/pathology
Collapse
Affiliation(s)
- Franziska Werner
- Institute of Physiology, University Würzburg, 97070 Würzburg, Germany; (F.W.); (T.N.); (L.S.); (K.V.); (L.K.)
| | - Takashi Naruke
- Institute of Physiology, University Würzburg, 97070 Würzburg, Germany; (F.W.); (T.N.); (L.S.); (K.V.); (L.K.)
| | - Lydia Sülzenbrück
- Institute of Physiology, University Würzburg, 97070 Würzburg, Germany; (F.W.); (T.N.); (L.S.); (K.V.); (L.K.)
| | - Sarah Schäfer
- Institute of Experimental Biomedicine, University Hospital Würzburg, 97080 Würzburg, Germany; (S.S.); (M.R.); (A.Z.)
| | - Melanie Rösch
- Institute of Experimental Biomedicine, University Hospital Würzburg, 97080 Würzburg, Germany; (S.S.); (M.R.); (A.Z.)
| | - Katharina Völker
- Institute of Physiology, University Würzburg, 97070 Würzburg, Germany; (F.W.); (T.N.); (L.S.); (K.V.); (L.K.)
| | - Lisa Krebes
- Institute of Physiology, University Würzburg, 97070 Würzburg, Germany; (F.W.); (T.N.); (L.S.); (K.V.); (L.K.)
| | - Marco Abeßer
- Institute of Physiology, University Würzburg, 97070 Würzburg, Germany; (F.W.); (T.N.); (L.S.); (K.V.); (L.K.)
| | - Dorothe Möllmann
- Institute of Pathology, University Hospital Essen, 45147 Essen, Germany; (D.M.); (H.A.B.)
| | - Hideo A. Baba
- Institute of Pathology, University Hospital Essen, 45147 Essen, Germany; (D.M.); (H.A.B.)
| | - Frank Schweda
- Institute of Physiology, University of Regensburg, 93053 Regensburg, Germany;
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, 97080 Würzburg, Germany; (S.S.); (M.R.); (A.Z.)
| | - Michaela Kuhn
- Institute of Physiology, University Würzburg, 97070 Würzburg, Germany; (F.W.); (T.N.); (L.S.); (K.V.); (L.K.)
| |
Collapse
|
19
|
Walzik D, Wences Chirino TY, Zimmer P, Joisten N. Molecular insights of exercise therapy in disease prevention and treatment. Signal Transduct Target Ther 2024; 9:138. [PMID: 38806473 PMCID: PMC11133400 DOI: 10.1038/s41392-024-01841-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/30/2024] Open
Abstract
Despite substantial evidence emphasizing the pleiotropic benefits of exercise for the prevention and treatment of various diseases, the underlying biological mechanisms have not been fully elucidated. Several exercise benefits have been attributed to signaling molecules that are released in response to exercise by different tissues such as skeletal muscle, cardiac muscle, adipose, and liver tissue. These signaling molecules, which are collectively termed exerkines, form a heterogenous group of bioactive substances, mediating inter-organ crosstalk as well as structural and functional tissue adaption. Numerous scientific endeavors have focused on identifying and characterizing new biological mediators with such properties. Additionally, some investigations have focused on the molecular targets of exerkines and the cellular signaling cascades that trigger adaption processes. A detailed understanding of the tissue-specific downstream effects of exerkines is crucial to harness the health-related benefits mediated by exercise and improve targeted exercise programs in health and disease. Herein, we review the current in vivo evidence on exerkine-induced signal transduction across multiple target tissues and highlight the preventive and therapeutic value of exerkine signaling in various diseases. By emphasizing different aspects of exerkine research, we provide a comprehensive overview of (i) the molecular underpinnings of exerkine secretion, (ii) the receptor-dependent and receptor-independent signaling cascades mediating tissue adaption, and (iii) the clinical implications of these mechanisms in disease prevention and treatment.
Collapse
Affiliation(s)
- David Walzik
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Tiffany Y Wences Chirino
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Philipp Zimmer
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
| | - Niklas Joisten
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
- Division of Exercise and Movement Science, Institute for Sport Science, University of Göttingen, 37075, Göttingen, Lower Saxony, Germany.
| |
Collapse
|
20
|
Dabaghie D, Charrin E, Tonelius P, Rosengren B, Korkut G, Granqvist AB, Lal M, Patrakka J. Unraveling the role of natriuretic peptide clearance receptor (NPR3) in glomerular diseases. Sci Rep 2024; 14:11850. [PMID: 38782980 PMCID: PMC11116399 DOI: 10.1038/s41598-024-61603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Natriuretic peptides (NPs) are cardio-derived hormones that have a crucial role in maintaining cardiovascular homeostasis. Physiological effects of NPs are mediated by binding to natriuretic peptide receptors 1 and 2 (NPR1/2), whereas natriuretic peptide receptor 3 (NPR3) acts as a clearance receptor that removes NPs from the circulation. Mouse studies have shown that local NP-signaling in the kidney glomerulus is important for the maintenance of renal homeostasis. In this study we examined the expression of NPR3 in kidney tissue and explored its involvement in renal physiology and disease by generating podocyte-specific knockout mice (NPR3podKO) as well as by using an NPR3 inhibitor (NPR3i) in rodent models of kidney disease. NPR3 was highly expressed by podocytes. NPR3podKO animals showed no renal abnormalities under healthy conditions and responded similarly to nephrotoxic serum (NTS) induced glomerular injury. However, NPR3i showed reno-protective effects in the NTS-induced model evidenced by decreased glomerulosclerosis and reduced podocyte loss. In a ZSF1 rat model of diabetic kidney injury, therapy alone with NPR3i did not have beneficial effects on renal function/histology, but when combined with losartan (angiotensin receptor blocker), NPR3i potentiated its ameliorative effects on albuminuria. In conclusion, these results suggest that NPR3 may contribute to kidney disease progression.
Collapse
Affiliation(s)
- Dina Dabaghie
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Emmanuelle Charrin
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Pernilla Tonelius
- Bioscience Renal, Cardiovascular, Renal and Metabolism (CVRM), R&D Biopharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Birgitta Rosengren
- Bioscience Renal, Cardiovascular, Renal and Metabolism (CVRM), R&D Biopharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Gizem Korkut
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Anna B Granqvist
- Bioscience Renal, Cardiovascular, Renal and Metabolism (CVRM), R&D Biopharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Mark Lal
- Bioscience Renal, Cardiovascular, Renal and Metabolism (CVRM), R&D Biopharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Jaakko Patrakka
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.
- Department of Pathology, Unilabs, Stockholm, Sweden.
| |
Collapse
|
21
|
Juraver-Geslin H, Devotta A, Saint-Jeannet JP. Developmental roles of natriuretic peptides and their receptors. Cells Dev 2023; 176:203878. [PMID: 37742795 PMCID: PMC10841480 DOI: 10.1016/j.cdev.2023.203878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023]
Abstract
Natriuretic peptides and their receptors are implicated in the physiological control of blood pressure, bone growth, and cardiovascular and renal homeostasis. They mediate their action through the modulation of intracellular levels of cGMP and cAMP, two second-messengers that have broad biological roles. In this review, we briefly describe the major players of this signaling pathway and their physiological roles in the adult, and discuss several reports describing their activity in the control of various aspects of embryonic development in several species. While the core components of this signaling pathway are well conserved, their functions have diverged in the embryo and the adult to control a diverse array of biological processes.
Collapse
Affiliation(s)
- Hugo Juraver-Geslin
- Department of Molecular Pathobiology, New York University, College of Dentistry, New York, NY 10010, USA
| | - Arun Devotta
- Department of Molecular Pathobiology, New York University, College of Dentistry, New York, NY 10010, USA
| | - Jean-Pierre Saint-Jeannet
- Department of Molecular Pathobiology, New York University, College of Dentistry, New York, NY 10010, USA.
| |
Collapse
|
22
|
Lu YY, Li SJ, Zhang Z, He S, Guo YT, Hong MN, Shao S, Wang RQ, Zhang J, Wang JG, Gao PJ, Li XD. C-atrial natriuretic peptide (ANP) 4-23 attenuates renal fibrosis in deoxycorticosterone-acetate-salt hypertensive mice. Exp Cell Res 2023; 431:113738. [PMID: 37572787 DOI: 10.1016/j.yexcr.2023.113738] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/08/2023] [Accepted: 08/04/2023] [Indexed: 08/14/2023]
Abstract
Epithelial-mesenchymal transition (EMT) plays a critical role in hypertension-induced renal fibrosis, a final pathway that leads to end-stage renal failure. C-Atrial natriuretic peptide (ANP)4-23, a specific agonist of natriuretic peptide receptor-C (NPR-C), has been reported to have protective effects against hypertension. However, the role of C-ANP4-23 in hypertension-associated renal fibrosis has not yet been elucidated. In this study, mice were randomly divided into SHAM group, DOCA-salt group and DOCA-salt + C-ANP4-23 group. Renal morphology changes, renal function and fibrosis were detected. Human proximal tubular epithelial cells (HK2) stimulated by aldosterone were used for cell function and mechanism study. The DOCA-salt treated mice exhibited hypertension, kidney fibrosis and renal dysfunction, which were attenuated by C-ANP4-23. Moreover, C-ANP4-23 inhibited DOCA-salt treatment-induced renal EMT as evidenced by decrease of the mesenchymal marker alpha-smooth muscle actin (ACTA2) and vimentin and increase of epithelial cell marker E-cadherin. In HK2 cells, aldosterone induced EMT response, which was also suppressed by C-ANP4-23. The key transcription factors (twist, snail, slug and ZEB1) involved in EMT were increased in the kidney of DOCA-salt-treated mice, which were also suppressed by C-ANP4-23. Mechanistically, C-ANP4-23 inhibited the aldosterone-induced translocation of MR from cytosol to nucleus without change of MR expression. Furthermore, C-ANP4-23 rescued the enhanced expression of NADPH oxidase (NOX) 4 and oxidative stress after aldosterone stimulation. Aldosterone-induced Akt and Erk1/2 activation was also suppressed by C-ANP4-23. Our data suggest that C-ANP4-23 attenuates renal fibrosis, likely through inhibition of MR activation, enhanced oxidative stress and Akt and Erk1/2 signaling pathway.
Collapse
Affiliation(s)
- Yuan-Yuan Lu
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China; Shanghai Geriatric Medical Center, Shanghai, China; Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shi-Jin Li
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China; State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Zhong Zhang
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Shun He
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Yue-Tong Guo
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Mo-Na Hong
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Shuai Shao
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Rui-Qi Wang
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jia Zhang
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Ji-Guang Wang
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Xiao-Dong Li
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| |
Collapse
|
23
|
Cheng C, Zhang J, Li X, Xue F, Cao L, Meng L, Sui W, Zhang M, Zhao Y, Xi B, Yu X, Xu F, Yang J, Zhang Y, Zhang C. NPRC deletion mitigated atherosclerosis by inhibiting oxidative stress, inflammation and apoptosis in ApoE knockout mice. Signal Transduct Target Ther 2023; 8:290. [PMID: 37553374 PMCID: PMC10409771 DOI: 10.1038/s41392-023-01560-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/24/2023] [Accepted: 07/07/2023] [Indexed: 08/10/2023] Open
Abstract
Previous studies suggested a beneficial effect of natriuretic peptides in animal models of cardiovascular disease, but the role of natriuretic peptide receptor C (NPRC) in the pathogenesis of atherosclerosis (AS) remains unknown. This study was designed to test the hypothesis that NPRC may promote AS lesion formation and instability by enhancing oxidative stress, inflammation, and apoptosis via protein kinase A (PKA) signaling. ApoE-/- mice were fed chow or Western diet for 12 weeks and NPRC expression was significantly increased in the aortic tissues of Western diet-fed mice. Systemic NPRC knockout mice were crossed with ApoE-/- mice to generate ApoE-/-NPRC-/- mice, and NPRC deletion resulted in a significant decrease in the size and instability of aortic atherosclerotic lesions in ApoE-/-NPRC-/- versus ApoE-/- mice. In addition, endothelial cell-specific NPRC knockout attenuated atherosclerotic lesions in mice. In contrast, endothelial cell overexpression of NPRC aggravated the size and instability of atherosclerotic aortic lesions in mice. Experiments in vitro showed that NPRC knockdown in human aortic endothelial cells (HAECs) inhibited ROS production, pro-inflammatory cytokine expression and endothelial cell apoptosis, and increased eNOS expression. Furthermore, NPRC knockdown in HAECs suppressed macrophage migration, cytokine expression, and phagocytosis via its effects on endothelial cells. On the contrary, NPRC overexpression in endothelial cells resulted in opposite effects. Mechanistically, the anti-inflammation and anti-atherosclerosis effects of NPRC deletion involved activation of cAMP/PKA pathway, leading to downstream upregulated AKT1 pathway and downregulated NF-κB pathway. In conclusion, NPRC deletion reduced the size and instability of atherosclerotic lesions in ApoE-/- mice via attenuating inflammation and endothelial cell apoptosis and increasing eNOS expression by modulating cAMP/PKA-AKT1 and NF-κB pathways. Thus, targeting NPRC may provide a promising approach to the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Cheng Cheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Jie Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaodong Li
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Fei Xue
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lei Cao
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Linlin Meng
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenhai Sui
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Meng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuxia Zhao
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Traditional Chinese Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Bo Xi
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feng Xu
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Jianmin Yang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Yun Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Cheng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
24
|
Meng L, Lu Y, Wang X, Cheng C, Xue F, Xie L, Zhang Y, Sui W, Zhang M, Zhang Y, Zhang C. NPRC deletion attenuates cardiac fibrosis in diabetic mice by activating PKA/PKG and inhibiting TGF-β1/Smad pathways. SCIENCE ADVANCES 2023; 9:eadd4222. [PMID: 37531438 PMCID: PMC10396312 DOI: 10.1126/sciadv.add4222] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/29/2023] [Indexed: 08/04/2023]
Abstract
Cardiac fibrosis plays a key role in the progression of diabetic cardiomyopathy (DCM). Previous studies demonstrated the cardioprotective effects of natriuretic peptides. However, the effects of natriuretic peptide receptor C (NPRC) on cardiac fibrosis in DCM remains unknown. Here, we observed that myocardial NPRC expression was increased in mice and patients with DCM. NPRC-/- diabetic mice showed alleviated cardiac fibrosis, as well as improved cardiac function and remodeling. NPRC knockdown in both cardiac fibroblasts and cardiomyocytes decreased collagen synthesis and proliferation of cardiac fibroblasts. RNA sequencing identified that NPRC deletion up-regulated the expression of TGF-β-induced factor homeobox 1 (TGIF1), which inhibited the phosphorylation of Smad2/3. Furthermore, TGIF1 up-regulation was mediated by the activation of cAMP/PKA and cGMP/PKG signaling induced by NPRC deletion. These findings suggest that NPRC deletion attenuated cardiac fibrosis and improved cardiac remodeling and function in diabetic mice, providing a promising approach to the treatment of diabetic cardiac fibrosis.
Collapse
Affiliation(s)
- Linlin Meng
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yue Lu
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xinlu Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Cheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Fei Xue
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Xie
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yaoyuan Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenhai Sui
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | | | - Yun Zhang
- Corresponding author. (Y.Z.); (C.Z.)
| | | |
Collapse
|
25
|
Liu D, Ceddia RP, Zhang W, Shi F, Fang H, Collins S. Discovery of another mechanism for the inhibition of particulate guanylyl cyclases by the natriuretic peptide clearance receptor. Proc Natl Acad Sci U S A 2023; 120:e2307882120. [PMID: 37399424 PMCID: PMC10334801 DOI: 10.1073/pnas.2307882120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 05/30/2023] [Indexed: 07/05/2023] Open
Abstract
The cardiac natriuretic peptides (NPs) control pivotal physiological actions such as fluid and electrolyte balance, cardiovascular homeostasis, and adipose tissue metabolism by activating their receptor enzymes [natriuretic peptide receptor-A (NPRA) and natriuretic peptide receptor-B (NPRB)]. These receptors are homodimers that generate intracellular cyclic guanosine monophosphate (cGMP). The natriuretic peptide receptor-C (NPRC), nicknamed the clearance receptor, lacks a guanylyl cyclase domain; instead, it can bind the NPs to internalize and degrade them. The conventional paradigm is that by competing for and internalizing NPs, NPRC blunts the ability of NPs to signal through NPRA and NPRB. Here we show another previously unknown mechanism by which NPRC can interfere with the cGMP signaling function of the NP receptors. By forming a heterodimer with monomeric NPRA or NPRB, NPRC can prevent the formation of a functional guanylyl cyclase domain and thereby suppress cGMP production in a cell-autonomous manner.
Collapse
Affiliation(s)
- Dianxin Liu
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, NashvilleTN37232
| | - Ryan P. Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, NashvilleTN37232
| | - Wei Zhang
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, NashvilleTN37232
| | - Fubiao Shi
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, NashvilleTN37232
| | - Huafeng Fang
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL32827
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, NashvilleTN37232
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN37232
| |
Collapse
|
26
|
Guarino BD, Dado CD, Kumar A, Braza J, Harrington EO, Klinger JR. Deletion of the Npr3 gene increases severity of acute lung injury in obese mice. Pulm Circ 2023; 13:e12270. [PMID: 37528869 PMCID: PMC10387407 DOI: 10.1002/pul2.12270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 08/03/2023] Open
Abstract
Previous studies have shown that atrial natriuretic peptide (ANP) attenuates agonist-induced pulmonary edema and that this effect may be mediated in part by the ANP clearance receptor, natriuretic peptide receptor-C (NPR-C). Obesity has been associated with lower plasma ANP levels due to increased expression of NPR-C, and with decreased severity of acute lung injury (ALI). Therefore, we hypothesized that increased expression of NPR-C may attenuate ALI severity in obese populations. To test this, we examined ALI in Npr3 wild-type (WT) and knockout (KO) mice fed normal chow (NC) or high-fat diets (HFD). After 12 weeks, ALI was induced with intra-tracheal administration of Pseudomonas aeruginosa strain 103 (PA103) or saline. ALI severity was determined by lung wet-to-dry ratio (W/D) along with measurement of cell count, protein levels from bronchoalveolar lavage fluid (BALF), and quantitative polymerase chain reaction was performed on whole lung to measure cytokine/chemokine and Npr3 mRNA expression. ANP levels were measured from plasma. PA103 caused ALI as determined by significant increases in W/D, BALF protein concentration, and whole lung cytokine/chemokine expression. PA103 increased Npr3 expression in the lungs of wild-type (WT) mice regardless of diet. There was a nonsignificant trend toward increased Npr3 expression in the lungs of WT mice fed HFD versus NC. No differences in ALI were seen between Npr3 knockout (KO) mice and WT-fed NC, but Npr3 KO mice fed HFD had a significantly greater W/D and BALF protein concentration than WT mice fed HFD. These findings support the hypothesis that Npr3 may help protect against ALI in obesity.
Collapse
Affiliation(s)
- Brianna D. Guarino
- Vascular Research LabProvidence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
- Department of Medicine, Sleep and Critical Care MedicineRhode Island HospitalProvidenceRhode IslandUSA
- Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Christopher D. Dado
- Vascular Research LabProvidence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
- Department of Medicine, Sleep and Critical Care MedicineRhode Island HospitalProvidenceRhode IslandUSA
- Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Ashok Kumar
- Vascular Research LabProvidence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
- Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Julie Braza
- Vascular Research LabProvidence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
- Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Elizabeth O. Harrington
- Vascular Research LabProvidence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
- Department of Medicine, Sleep and Critical Care MedicineRhode Island HospitalProvidenceRhode IslandUSA
- Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - James R. Klinger
- Vascular Research LabProvidence Veterans Affairs Medical CenterProvidenceRhode IslandUSA
- Department of Medicine, Sleep and Critical Care MedicineRhode Island HospitalProvidenceRhode IslandUSA
- Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| |
Collapse
|
27
|
Della Corte V, Pacinella G, Todaro F, Pecoraro R, Tuttolomondo A. The Natriuretic Peptide System: A Single Entity, Pleiotropic Effects. Int J Mol Sci 2023; 24:ijms24119642. [PMID: 37298592 DOI: 10.3390/ijms24119642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
In the modern scientific landscape, natriuretic peptides are a complex and interesting network of molecules playing pleiotropic effects on many organs and tissues, ensuring the maintenance of homeostasis mainly in the cardiovascular system and regulating the water-salt balance. The characterization of their receptors, the understanding of the molecular mechanisms through which they exert their action, and the discovery of new peptides in the last period have made it possible to increasingly feature the physiological and pathophysiological role of the members of this family, also allowing to hypothesize the possible settings for using these molecules for therapeutic purposes. This literature review traces the history of the discovery and characterization of the key players among the natriuretic peptides, the scientific trials performed to ascertain their physiological role, and the applications of this knowledge in the clinical field, leaving a glimpse of new and exciting possibilities for their use in the treatment of diseases.
Collapse
Affiliation(s)
- Vittoriano Della Corte
- Internal Medicine and Stroke Care Ward, Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialities (PROMISE) "G. D'Alessandro", University of Palermo, 90127 Palermo, Italy
| | - Gaetano Pacinella
- Internal Medicine and Stroke Care Ward, Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialities (PROMISE) "G. D'Alessandro", University of Palermo, 90127 Palermo, Italy
| | - Federica Todaro
- Internal Medicine and Stroke Care Ward, Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialities (PROMISE) "G. D'Alessandro", University of Palermo, 90127 Palermo, Italy
| | - Rosaria Pecoraro
- Internal Medicine and Stroke Care Ward, Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialities (PROMISE) "G. D'Alessandro", University of Palermo, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Internal Medicine and Stroke Care Ward, Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialities (PROMISE) "G. D'Alessandro", University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
28
|
González-Prendes R, Derks MFL, Groenen MAM, Quintanilla R, Amills M. Assessing the relationship between the in silico predicted consequences of 97 missense mutations mapping to 68 genes related to lipid metabolism and their association with porcine fatness traits. Genomics 2023; 115:110589. [PMID: 36842749 DOI: 10.1016/j.ygeno.2023.110589] [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: 08/20/2022] [Revised: 01/29/2023] [Accepted: 02/21/2023] [Indexed: 02/26/2023]
Abstract
In general, the relationship between the predicted functional consequences of missense mutations mapping to genes known to be involved in human diseases and the severity of disease manifestations is weak. In this study, we tested in pigs whether missense single nucleotide polymorphisms (SNPs), predicted to have consequences on the function of genes related to lipid metabolism are associated with lipid phenotypes. Association analysis demonstrated that nine out of 72 nominally associated SNPs were classified as "highly" or "very highly consistent" in silico-predicted functional mutations and did not show association with lipid traits expected to be affected by inactivation of the corresponding gene. Although the lack of endophenotypes and the limited sample size of certain genotypic classes might have limited to some extent the reach of the current study, our data indicate that present-day bioinformatic tools have a modest ability to predict the impact of missense mutations on complex phenotypes.
Collapse
Affiliation(s)
- Rayner González-Prendes
- Animal Breeding and Genomics, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, the Netherlands.
| | - Martijn F L Derks
- Animal Breeding and Genomics, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, the Netherlands
| | - Martien A M Groenen
- Animal Breeding and Genomics, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, the Netherlands
| | - Raquel Quintanilla
- Animal Breeding and Genetics Program, Institute of Agrifood Research and Technology (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | - Marcel Amills
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| |
Collapse
|
29
|
Pandey KN. Guanylyl cyclase/natriuretic peptide receptor-A: Identification, molecular characterization, and physiological genomics. Front Mol Neurosci 2023; 15:1076799. [PMID: 36683859 PMCID: PMC9846370 DOI: 10.3389/fnmol.2022.1076799] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/02/2022] [Indexed: 01/06/2023] Open
Abstract
The natriuretic peptides (NPs) hormone family, which consists mainly of atrial, brain, and C-type NPs (ANP, BNP, and CNP), play diverse roles in mammalian species, ranging from renal, cardiac, endocrine, neural, and vascular hemodynamics to metabolic regulations, immune responsiveness, and energy distributions. Over the last four decades, new data has transpired regarding the biochemical and molecular compositions, signaling mechanisms, and physiological and pathophysiological functions of NPs and their receptors. NPs are incremented mainly in eliciting natriuretic, diuretic, endocrine, vasodilatory, and neurological activities, along with antiproliferative, antimitogenic, antiinflammatory, and antifibrotic responses. The main locus responsible in the biological and physiological regulatory actions of NPs (ANP and BNP) is the plasma membrane guanylyl cyclase/natriuretic peptide receptor-A (GC-A/NPRA), a member of the growing multi-limbed GC family of receptors. Advances in this field have provided tremendous insights into the critical role of Npr1 (encoding GC-A/NPRA) in the reduction of fluid volume and blood pressure homeostasis, protection against renal and cardiac remodeling, and moderation and mediation of neurological disorders. The generation and use of genetically engineered animals, including gene-targeted (gene-knockout and gene-duplication) and transgenic mutant mouse models has revealed and clarified the varied roles and pleiotropic functions of GC-A/NPRA in vivo in intact animals. This review provides a chronological development of the biochemical, molecular, physiological, and pathophysiological functions of GC-A/NPRA, including signaling pathways, genomics, and gene regulation in both normal and disease states.
Collapse
|
30
|
Miller KJ, Henry I, Maylin Z, Smith C, Arunachalam E, Pandha H, Asim M. A compendium of Androgen Receptor Variant 7 target genes and their role in Castration Resistant Prostate Cancer. Front Oncol 2023; 13:1129140. [PMID: 36937454 PMCID: PMC10014620 DOI: 10.3389/fonc.2023.1129140] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/13/2023] [Indexed: 03/05/2023] Open
Abstract
Persistent androgen receptor (AR) signalling is the main driver of prostate cancer (PCa). Truncated isoforms of the AR called androgen receptor variants (AR-Vs) lacking the ligand binding domain often emerge during treatment resistance against AR pathway inhibitors such as Enzalutamide. This review discusses how AR-Vs drive a more aggressive form of PCa through the regulation of some of their target genes involved in oncogenic pathways, enabling disease progression. There is a pressing need for the development of a new generation of AR inhibitors which can repress the activity of both the full-length AR and AR-Vs, for which the knowledge of differentially expressed target genes will allow evaluation of inhibition efficacy. This review provides a detailed account of the most common variant, AR-V7, the AR-V7 regulated genes which have been experimentally validated, endeavours to understand their relevance in aggressive AR-V driven PCa and discusses the utility of the downstream protein products as potential drug targets for PCa treatment.
Collapse
Affiliation(s)
| | | | - Zoe Maylin
- *Correspondence: Zoe Maylin, ; Mohammad Asim,
| | | | | | | | | |
Collapse
|
31
|
Lu G, Hu R, Tao T, Hu M, Dong Z, Wang C. Regulatory role of atrial natriuretic peptide in brown adipose tissue: A narrative review. Obes Rev 2023; 24:e13522. [PMID: 36336901 DOI: 10.1111/obr.13522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/15/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022]
Abstract
Atrial natriuretic peptide (ANP) has been considered to exert an essential role as a cardiac secretory hormone in the regulation of hemodynamic homeostasis. As the research progresses, the role of ANP in the crosstalk between heart and lipid metabolism has become an interesting topic that is attracting the interest of researchers. The regulation of ANP in lipid metabolism shows favorable effects, particularly the activation of brown adipose tissue (BAT). The complex regulatory network of ANP on BAT has not been fully outlined. This narrative review critically evaluated the existing literature on the regulatory effects of ANP on BAT. In general, we have summarized the expression of ANP and its receptors in various human tissues, analyzed the progress of research on the relationship between the ANP and BAT, and described several potential pathways of ANP to BAT. Exogenous ANP, natriuretic peptide receptor C (NPRC) deficiency, cold exposure, bariatric surgery, and cardiac or renal insufficiency could all contribute to BAT expression by increasing circulating ANP levels.
Collapse
Affiliation(s)
- Guanhua Lu
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
- Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| | - Ruixiang Hu
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
- Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| | - Tian Tao
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
- Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| | - Min Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Zhiyong Dong
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
- Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
- Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
32
|
Heinl ES, Broeker KAE, Lehrmann C, Heydn R, Krieger K, Ortmaier K, Tauber P, Schweda F. Localization of natriuretic peptide receptors A, B, and C in healthy and diseased mouse kidneys. Pflugers Arch 2023; 475:343-360. [PMID: 36480070 PMCID: PMC9908653 DOI: 10.1007/s00424-022-02774-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022]
Abstract
The natriuretic peptides (NPs) ANP (atrial natriuretic peptide) and BNP (B-type natriuretic peptide) mediate their widespread effects by activating the natriuretic peptide receptor-A (NPR-A), while C-type natriuretic peptide (CNP) acts via natriuretic peptide receptor-B (NPR-B). NPs are removed from the circulation by internalization via the natriuretic peptide clearance receptor natriuretic peptide receptor-C (NPR-C). In addition to their well-known functions, for instance on blood pressure, all three NPs confer significant cardioprotection and renoprotection. Since neither the NP-mediated renal functions nor the renal target cells of renoprotection are completely understood, we performed systematic localization studies of NP receptors using in situ hybridization (RNAscope) in mouse kidneys. NPR-A mRNA is highly expressed in glomeruli (mainly podocytes), renal arterioles, endothelial cells of peritubular capillaries, and PDGFR-receptor β positive (PDGFR-β) interstitial cells. No NPR-A mRNA was detected by RNAscope in the tubular system. In contrast, NPR-B expression is highest in proximal tubules. NPR-C is located in glomeruli (mainly podocytes), in endothelial cells and PDGFR-β positive cells. To test for a possible regulation of NPRs in kidney diseases, their distribution was studied in adenine nephropathy. Signal intensity of NPR-A and NPR-B mRNA was reduced while their spatial distribution was unaltered compared with healthy kidneys. In contrast, NPR-C mRNA signal was markedly enhanced in cell clusters of myofibroblasts in fibrotic areas of adenine kidneys. In conclusion, the primary renal targets of ANP and BNP are glomerular, vascular, and interstitial cells but not the tubular compartment, while the CNP receptor NPR-B is highly expressed in proximal tubules. Further studies are needed to clarify the function and interplay of this specific receptor expression pattern.
Collapse
Affiliation(s)
- Elena-Sofia Heinl
- Institute for Physiology, University Regensburg, Regensburg, Germany.
| | | | - Claudia Lehrmann
- grid.7727.50000 0001 2190 5763Institute for Physiology, University Regensburg, Regensburg, Germany
| | - Rosmarie Heydn
- grid.7727.50000 0001 2190 5763Institute for Physiology, University Regensburg, Regensburg, Germany
| | - Katharina Krieger
- grid.7727.50000 0001 2190 5763Institute for Physiology, University Regensburg, Regensburg, Germany
| | - Katharina Ortmaier
- grid.7727.50000 0001 2190 5763Institute for Physiology, University Regensburg, Regensburg, Germany
| | - Philipp Tauber
- grid.7727.50000 0001 2190 5763Institute for Physiology, University Regensburg, Regensburg, Germany
| | - Frank Schweda
- Institute for Physiology, University Regensburg, Regensburg, Germany.
| |
Collapse
|
33
|
Sarzani R, Allevi M, Di Pentima C, Schiavi P, Spannella F, Giulietti F. Role of Cardiac Natriuretic Peptides in Heart Structure and Function. Int J Mol Sci 2022; 23:ijms232214415. [PMID: 36430893 PMCID: PMC9697447 DOI: 10.3390/ijms232214415] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Cardiac natriuretic peptides (NPs), atrial NP (ANP) and B-type NP (BNP) are true hormones produced and released by cardiomyocytes, exerting several systemic effects. Together with C-type NP (CNP), mainly expressed by endothelial cells, they also exert several paracrine and autocrine activities on the heart itself, contributing to cardiovascular (CV) health. In addition to their natriuretic, vasorelaxant, metabolic and antiproliferative systemic properties, NPs prevent cardiac hypertrophy, fibrosis, arrhythmias and cardiomyopathies, counteracting the development and progression of heart failure (HF). Moreover, recent studies revealed that a protein structurally similar to NPs mainly produced by skeletal muscles and osteoblasts called musclin/osteocrin is able to interact with the NPs clearance receptor, attenuating cardiac dysfunction and myocardial fibrosis and promoting heart protection during pathological overload. This narrative review is focused on the direct activities of this molecule family on the heart, reporting both experimental and human studies that are clinically relevant for physicians.
Collapse
Affiliation(s)
- Riccardo Sarzani
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
- Correspondence: (R.S.); Tel.: +39-071-5964696
| | - Massimiliano Allevi
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Chiara Di Pentima
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
| | - Paola Schiavi
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Francesco Spannella
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Federico Giulietti
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
| |
Collapse
|
34
|
In Silico Study of the Mechanisms Underlying the Action of the Snake Natriuretic-Like Peptide Lebetin 2 during Cardiac Ischemia. Toxins (Basel) 2022; 14:toxins14110787. [PMID: 36422961 PMCID: PMC9699598 DOI: 10.3390/toxins14110787] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Lebetin 2 (L2), a natriuretic-like peptide (NP), exerts potent cardioprotection in myocardial infarction (MI), with stronger effects than B-type natriuretic peptide (BNP). To determine the molecular mechanisms underlying its cardioprotection effect, we used molecular modeling, molecular docking and molecular dynamics (MD) simulation to describe the binding mode, key interaction residues as well as mechanistic insights into L2 interaction with NP receptors (NPRs). L2 binding affinity was determined for human, rat, mouse and chicken NPRs, and the stability of receptor-ligand complexes ascertained during 100 ns-long MD simulations. We found that L2 exhibited higher affinity for all human NPRs compared to BNP, with a rank preference for NPR-A > NPR-C > NPR-B. Moreover, L2 affinity for human NPR-A and NPR-C was higher in other species. Both docking and MD studies revealed that the NPR-C-L2 interaction was stronger in all species compared to BNP. Due to its higher affinity to human receptors, L2 could be used as a therapeutic approach in MI patients. Moreover, the stronger interaction of L2 with NPR-C could highlight a new L2 signaling pathway that would explain its additional effects during cardiac ischemia. Thus, L2 is a promising candidate for drug design toward novel compounds with high potency, affinity and stability.
Collapse
|
35
|
Khurana ML, Mani I, Kumar P, Ramasamy C, Pandey KN. Ligand-Dependent Downregulation of Guanylyl Cyclase/Natriuretic Peptide Receptor-A: Role of miR-128 and miR-195. Int J Mol Sci 2022; 23:ijms232113381. [PMID: 36362173 PMCID: PMC9657974 DOI: 10.3390/ijms232113381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Cardiac hormones act on the regulation of blood pressure (BP) and cardiovascular homeostasis. These hormones include atrial and brain natriuretic peptides (ANP, BNP) and activate natriuretic peptide receptor-A (NPRA), which enhance natriuresis, diuresis, and vasorelaxation. In this study, we established the ANP-dependent homologous downregulation of NPRA using human embryonic kidney-293 (HEK-293) cells expressing recombinant receptor and MA-10 cells harboring native endogenous NPRA. The prolonged pretreatment of cells with ANP caused a time- and dose-dependent decrease in 125I-ANP binding, Guanylyl cyclase (GC) activity of receptor, and intracellular accumulation of cGMP leading to downregulation of NPRA. Treatment with ANP (100 nM) for 12 h led to an 80% decrease in 125I-ANP binding to its receptor, and BNP decreased it by 62%. Neither 100 nM c-ANF (truncated ANF) nor C-type natriuretic peptide (CNP) had any effect. ANP (100 nM) treatment also decreased GC activity by 68% and intracellular accumulation cGMP levels by 45%, while the NPRA antagonist A71915 (1 µM) almost completely blocked ANP-dependent downregulation of NPRA. Treatment with the protein kinase G (PKG) stimulator 8-(4-chlorophenylthio)-cGMP (CPT-cGMP) (1 µM) caused a significant increase in 125I-ANP binding, whereas the PKG inhibitor KT 5823 (1 µM) potentiated the effect of ANP on the downregulation of NPRA. The transfection of miR-128 significantly reduced NPRA protein levels by threefold compared to control cells. These results suggest that ligand-dependent mechanisms play important roles in the downregulation of NPRA in target cells.
Collapse
|
36
|
Harrington EO, Kumar A, Leandre V, Wilson ZS, Guarino B, Braza J, Lefort CT, Klinger JR. Natriuretic peptide receptor-C mediates the inhibitory effect of atrial natriuretic peptide on neutrophil recruitment to the lung during acute lung injury. Am J Physiol Lung Cell Mol Physiol 2022; 323:L438-L449. [PMID: 35943160 PMCID: PMC9529260 DOI: 10.1152/ajplung.00477.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 01/08/2023] Open
Abstract
Atrial natriuretic peptide (ANP) protects against acute lung injury (ALI), but the receptor that mediates this effect is not known. Transgenic mice with 0 (knockout), 1 (heterozygote), or 2 (wild-type) functional copies of Npr3, the gene that encodes for natriuretic peptide receptor-C (NPR-C), were treated with intravenous infusion of ANP or saline vehicle before oropharyngeal aspiration of Pseudomonas aeruginosa (PA103) or saline vehicle. Lung injury was assessed 4 h following aspiration by measurement of lung wet/dry (W/D) weight, whole lung leukocyte and cytokine levels, and protein, leukocyte, and cytokine concentration in bronchoalveolar lavage fluid (BALF). PA103 induced acute lung injury as evidenced by increases in lung W/D ratio and protein concentration in BALF. The severity of PA103-induced lung injury did not differ between NPR-C genotypes. Treatment with intravenous ANP infusion reduced PA103-induced increases in lung W/D and BALF protein concentration in all three NPRC genotypes. PA103 increased the percentage of leukocytes that were neutrophils and cytokine levels in whole lung and BALF in NPR-C wild-type and knockout mice. This effect was blunted by ANP in wild-type mice but not in the NPR-C knockout mice. NPR-C does not mediate the protective effect of ANP on endothelial cell permeability in settings of PA103-induced injury but may mediate the effect of ANP on inhibition of the recruitment of neutrophils to the lung and thereby attenuate the release of inflammatory cytokines.
Collapse
Affiliation(s)
- Elizabeth O Harrington
- Vascular Research Lab, Providence Veterans Administration Medical Center, Providence, Rhode Island
- Division of Pulmonary, Sleep and Critical Care Medicine, Rhode Island Hospital, Providence, Rhode Island
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Ashok Kumar
- Vascular Research Lab, Providence Veterans Administration Medical Center, Providence, Rhode Island
| | - Verida Leandre
- Pathobiology Graduate Program, Brown University, Providence, Rhode Island
| | - Zachary S Wilson
- Pathobiology Graduate Program, Brown University, Providence, Rhode Island
| | - Brianna Guarino
- Vascular Research Lab, Providence Veterans Administration Medical Center, Providence, Rhode Island
- Division of Pulmonary, Sleep and Critical Care Medicine, Rhode Island Hospital, Providence, Rhode Island
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Julie Braza
- Vascular Research Lab, Providence Veterans Administration Medical Center, Providence, Rhode Island
| | - Craig T Lefort
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Providence, Rhode Island
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - James R Klinger
- Vascular Research Lab, Providence Veterans Administration Medical Center, Providence, Rhode Island
- Division of Pulmonary, Sleep and Critical Care Medicine, Rhode Island Hospital, Providence, Rhode Island
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
37
|
Ang WF, Koh CY, Kini RM. From Snake Venoms to Therapeutics: A Focus on Natriuretic Peptides. Pharmaceuticals (Basel) 2022; 15:ph15091153. [PMID: 36145374 PMCID: PMC9502559 DOI: 10.3390/ph15091153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 11/26/2022] Open
Abstract
Snake venom is a cocktail of multifunctional biomolecules that has evolved with the purpose of capturing prey and for defense. These biomolecules are classified into different classes based on their functions. They include three-finger toxins, natriuretic peptides, phospholipases and metalloproteinases. The focus for this review is on the natriuretic peptide (NP), which is an active component that can be isolated from the venoms of vipers and mambas. In these venoms, NPs contribute to the lowering of blood pressure, causing a rapid loss of consciousness in the prey such that its mobility is reduced, paralyzing the prey, and often death follows. Over the past 30 years since the discovery of the first NP in the venom of the green mamba, venom NPs have shown potential in the development of drug therapy for heart failure. Venom NPs have long half-lives, different pharmacological profiles, and may also possess different functions in comparison to the mammalian NPs. Understanding their mechanisms of action provides the strategies needed to develop new NPs for treatment of heart failure. This review summarizes the venom NPs that have been identified over the years and how they can be useful in drug development.
Collapse
Affiliation(s)
- Wei Fong Ang
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117558, Singapore
- NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore 119077, Singapore
| | - Cho Yeow Koh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117559, Singapore
- Correspondence: (C.Y.K.); (R.M.K.); Tel.: +65-6601-1387 (C.Y.K.); +65-6516-5235 (R.M.K.)
| | - R. Manjunatha Kini
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117558, Singapore
- NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore 119077, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298-0614, USA
- Correspondence: (C.Y.K.); (R.M.K.); Tel.: +65-6601-1387 (C.Y.K.); +65-6516-5235 (R.M.K.)
| |
Collapse
|
38
|
Lauffer P, Boudin E, van der Kaay DCM, Koene S, van Haeringen A, van Tellingen V, Van Hul W, Prickett TCR, Mortier G, Espiner EA, van Duyvenvoorde HA. Broadening the Spectrum of Loss-of-Function Variants in NPR-C-Related Extreme Tall Stature. J Endocr Soc 2022; 6:bvac019. [PMID: 35233476 PMCID: PMC8879884 DOI: 10.1210/jendso/bvac019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Indexed: 11/28/2022] Open
Abstract
CONTEXT Natriuretic peptide receptor-C (NPR-C, encoded by NPR3) belongs to a family of cell membrane-integral proteins implicated in various physiological processes, including longitudinal bone growth. NPR-C acts as a clearance receptor of natriuretic peptides, including C-type natriuretic peptide (CNP), that stimulate the cGMP-forming guanylyl cyclase-coupled receptors NPR-A and NPR-B. Pathogenic variants in CNP, NPR2, and NPR3 may cause a tall stature phenotype associated with macrodactyly of the halluces and epiphyseal dysplasia. OBJECTIVE Here we report on a boy with 2 novel biallelic inactivating variants of NPR3. METHODS History and clinical characteristics were collected. Biochemical indices of natriuretic peptide clearance and in vitro cellular localization of NPR-C were studied to investigate causality of the identified variants. RESULTS We identified 2 novel compound heterozygous NPR3 variants c.943G>A p.(Ala315Thr) and c.1294A>T p.(Ile432Phe) in a boy with tall stature and macrodactyly of the halluces. In silico analysis indicated decreased stability of NPR-C, presumably resulting in increased degradation or trafficking defects. Compared to other patients with NPR-C loss-of-function, the phenotype seemed to be milder: pseudo-epiphyses in hands and feet were absent, biochemical features were less severe, and there was some co-localization of p.(Ile432Phe) NPR-C with the cell membrane, as opposed to complete cytoplasmic retention. CONCLUSION With this report on a boy with tall stature and macrodactyly of the halluces we further broaden the genotypic and phenotypic spectrum of NPR-C-related tall stature.
Collapse
Affiliation(s)
- Peter Lauffer
- Department of Pediatric Endocrinology, Emma Children’s Hospital, Amsterdam University Medical Center, 1105 AZ Amsterdam, the Netherlands
| | - Eveline Boudin
- Department of Medical Genetics, Antwerp University Hospital and University of Antwerp, 2650 Edegem, Belgium
| | - Daniëlle C M van der Kaay
- Department of Pediatric Endocrinology, Sophia Children’s Hospital, Erasmus Medical Center, 3015 GD Rotterdam, the Netherlands
| | - Saskia Koene
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Arie van Haeringen
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Vera van Tellingen
- Department of Pediatrics, Catharina Hospital, 5623 EJ Eindhoven, the Netherlands
| | - Wim Van Hul
- Department of Medical Genetics, Antwerp University Hospital and University of Antwerp, 2650 Edegem, Belgium
| | | | - Geert Mortier
- Department of Medical Genetics, Antwerp University Hospital and University of Antwerp, 2650 Edegem, Belgium
| | - Eric A Espiner
- Department of Medicine, University of Otago, 8140 Christchurch, New Zealand
| | | |
Collapse
|
39
|
Expanding horizons of achondroplasia treatment: current options and future developments. Osteoarthritis Cartilage 2022; 30:535-544. [PMID: 34864168 DOI: 10.1016/j.joca.2021.11.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/23/2021] [Accepted: 11/28/2021] [Indexed: 02/02/2023]
Abstract
Activating mutations in the FGFR3 receptor tyrosine kinase lead to most prevalent form of genetic dwarfism in humans, the achondroplasia. Many features of the complex function of FGFR3 in growing skeleton were characterized, which facilitated identification of therapy targets, and drove progress toward treatment. In August 2021, the vosoritide was approved for treatment of achondroplasia, which is based on a stable variant of the C-natriuretic peptide. Other drugs may soon follow, as several conceptually different inhibitors of FGFR3 signaling progress through clinical trials. Here, we review the current achondroplasia therapeutics, describe their mechanisms, and illuminate motivations leading to their development. We also discuss perspectives of curing achondroplasia, and options for repurposing achondroplasia drugs for dwarfing conditions unrelated to FGFR3.
Collapse
|
40
|
Abstract
The global mortality, morbidity, and healthcare costs associated with cardiometabolic disease, including obesity, diabetes, hypertension, and dyslipidemia, are substantial and represent an expanding unmet medical need. Herein, we have identified a physiological role for C-type natriuretic peptide (CNP) in regulating key processes, including thermogenesis and adipogenesis, which combine to coordinate metabolic function and prevent the development of cardiometabolic disorders. This protective mechanism, which is in part mediated via an autocrine action of CNP on adipocytes, is underpinned by activation of cognate natriuretic peptide receptors (NPR)-B and NPR-C. This mechanism advances the fundamental understanding of energy homeostasis and glucose handling and offers the promise of improving the treatment of cardiometabolic disease. Thermogenesis and adipogenesis are tightly regulated mechanisms that maintain lipid homeostasis and energy balance; dysfunction of these critical processes underpins obesity and contributes to cardiometabolic disease. C-type natriuretic peptide (CNP) fulfills a multimodal protective role in the cardiovascular system governing local blood flow, angiogenesis, cardiac function, and immune cell reactivity. Herein, we investigated a parallel, preservative function for CNP in coordinating metabolic homeostasis. Global inducible CNP knockout mice exhibited reduced body weight, higher temperature, lower adiposity, and greater energy expenditure in vivo. This thermogenic phenotype was associated with increased expression of uncoupling protein-1 and preferential lipid utilization by mitochondria, a switch corroborated by a corresponding diminution of insulin secretion and glucose clearance. Complementary studies in isolated murine and human adipocytes revealed that CNP exerts these metabolic regulatory actions by inhibiting sympathetic thermogenic programming via Gi-coupled natriuretic peptide receptor (NPR)-C and reducing peroxisome proliferator-activated receptor-γ coactivator-1α expression, while concomitantly driving adipogenesis via NPR-B/protein kinase-G. Finally, we identified an association between CNP/NPR-C expression and obesity in patient samples. These findings establish a pivotal physiological role for CNP as a metabolic switch to balance energy homeostasis. Pharmacological targeting of these receptors may offer therapeutic utility in the metabolic syndrome and related cardiovascular disorders.
Collapse
|
41
|
Saxena A, Sharma V, Muthuirulan P, Neufeld SJ, Tran MP, Gutierrez HL, Chen KD, Erberich JM, Birmingham A, Capellini TD, Cobb J, Hiller M, Cooper KL. Interspecies transcriptomics identify genes that underlie disproportionate foot growth in jerboas. Curr Biol 2022; 32:289-303.e6. [PMID: 34793695 PMCID: PMC8792248 DOI: 10.1016/j.cub.2021.10.063] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 07/16/2021] [Accepted: 10/28/2021] [Indexed: 01/26/2023]
Abstract
Despite the great diversity of vertebrate limb proportion and our deep understanding of the genetic mechanisms that drive skeletal elongation, little is known about how individual bones reach different lengths in any species. Here, we directly compare the transcriptomes of homologous growth cartilages of the mouse (Mus musculus) and bipedal jerboa (Jaculus jaculus), the latter of which has "mouse-like" arms but extremely long metatarsals of the feet. Intersecting gene-expression differences in metatarsals and forearms of the two species revealed that about 10% of orthologous genes are associated with the disproportionately rapid elongation of neonatal jerboa feet. These include genes and enriched pathways not previously associated with endochondral elongation as well as those that might diversify skeletal proportion in addition to their known requirements for bone growth throughout the skeleton. We also identified transcription regulators that might act as "nodes" for sweeping differences in genome expression between species. Among these, Shox2, which is necessary for proximal limb elongation, has gained expression in jerboa metatarsals where it has not been detected in other vertebrates. We show that Shox2 is sufficient to increase mouse distal limb length, and a nearby putative cis-regulatory region is preferentially accessible in jerboa metatarsals. In addition to mechanisms that might directly promote growth, we found evidence that jerboa foot elongation may occur in part by de-repressing latent growth potential. The genes and pathways that we identified here provide a framework to understand the modular genetic control of skeletal growth and the remarkable malleability of vertebrate limb proportion.
Collapse
Affiliation(s)
- Aditya Saxena
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Virag Sharma
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, Dresden 01307, Germany; Max Planck Institute for the Physics of Complex Systems, Nothnitzerstraße 38, Dresden 01187, Germany
| | - Pushpanathan Muthuirulan
- Department of Human Evolutionary Biology, Harvard University, 11 Divinity Avenue, Cambridge, MA 02138, USA
| | - Stanley J Neufeld
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Mai P Tran
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Haydee L Gutierrez
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kevin D Chen
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Joel M Erberich
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Amanda Birmingham
- Center for Computational Biology and Bioinformatics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, 11 Divinity Avenue, Cambridge, MA 02138, USA
| | - John Cobb
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Michael Hiller
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, Dresden 01307, Germany; Max Planck Institute for the Physics of Complex Systems, Nothnitzerstraße 38, Dresden 01187, Germany
| | - Kimberly L Cooper
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
42
|
Wagner BM, Robinson JW, Healy CL, Gauthier M, Dickey DM, Yee SP, Osborn JW, O’Connell TD, Potter LR. Guanylyl cyclase-A phosphorylation decreases cardiac hypertrophy and improves systolic function in male, but not female, mice. FASEB J 2022; 36:e22069. [PMID: 34859913 PMCID: PMC8826535 DOI: 10.1096/fj.202100600rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 01/03/2023]
Abstract
Atrial natriuretic peptide (NP) and BNP increase cGMP, which reduces blood pressure and cardiac hypertrophy by activating guanylyl cyclase (GC)-A, also known as NPR-A or Npr1. Although GC-A is highly phosphorylated, and dephosphorylation inactivates the enzyme, the significance of GC-A phosphorylation to heart structure and function remains unknown. To identify in vivo processes that are regulated by GC-A phosphorylation, we substituted glutamates for known phosphorylation sites to make GC-A8E/8E mice that express an enzyme that cannot be inactivated by dephosphorylation. GC-A activity, but not protein, was increased in heart and kidney membranes from GC-A8E/8E mice. Activities were threefold higher in female compared to male cardiac ventricles. Plasma cGMP and testosterone were elevated in male and female GC-A8E/8E mice, but aldosterone was only increased in mutant male mice. Plasma and urinary creatinine concentrations were decreased and increased, respectively, but blood pressure and heart rate were unchanged in male GC-A8E/8E mice. Heart weight to body weight ratios for GC-A8E/8E male, but not female, mice were 12% lower with a 14% reduction in cardiomyocyte cross-sectional area. Subcutaneous injection of fsANP, a long-lived ANP analog, increased plasma cGMP and decreased aldosterone in male GC-AWT/WT and GC-A8E/8E mice at 15 min, but only GC-A8E/8E mice had elevated levels of plasma cGMP and aldosterone at 60 min. fsANP reduced ventricular ERK1/2 phosphorylation to a greater extent and for a longer time in the male mutant compared to WT mice. Finally, ejection fractions were increased in male but not female hearts from GC-A8E/8E mice. We conclude that increased phosphorylation-dependent GC-A activity decreases cardiac ERK activity, which results in smaller male hearts with improved systolic function.
Collapse
Affiliation(s)
- Brandon M. Wagner
- Department of Integrative Biology and Physiology, University of Minnesota, Medical School, Minneapolis, MN 55455 USA
| | - Jerid W. Robinson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Medical School, Minneapolis, MN 55455 USA
| | - Chastity L. Healy
- Department of Integrative Biology and Physiology, University of Minnesota, Medical School, Minneapolis, MN 55455 USA
| | - Madeline Gauthier
- Department of Integrative Biology and Physiology, University of Minnesota, Medical School, Minneapolis, MN 55455 USA
| | - Deborah M. Dickey
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Medical School, Minneapolis, MN 55455 USA
| | - Siu-Pok Yee
- Department of Cell Biology at the University of Connecticut Health Center, Farmington, CT 06030 USA
| | - John W. Osborn
- Department of Surgery at the University of Minnesota, Medical School, Minneapolis, MN 55455 USA
| | - Timothy D. O’Connell
- Department of Integrative Biology and Physiology, University of Minnesota, Medical School, Minneapolis, MN 55455 USA,,Corresponding authors: Timothy D O’Connell , Lincoln R Potter
| | - Lincoln R. Potter
- Department of Integrative Biology and Physiology, University of Minnesota, Medical School, Minneapolis, MN 55455 USA,,Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Medical School, Minneapolis, MN 55455 USA,,Corresponding authors: Timothy D O’Connell , Lincoln R Potter
| |
Collapse
|
43
|
Takei Y. Evolution of the membrane/particulate guanylyl cyclase: From physicochemical sensors to hormone receptors. Gen Comp Endocrinol 2022; 315:113797. [PMID: 33957096 DOI: 10.1016/j.ygcen.2021.113797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 12/26/2022]
Abstract
Guanylyl cyclase (GC) is an enzyme that produces 3',5'-cyclic guanosine monophosphate (cGMP), one of the two canonical cyclic nucleotides used as a second messenger for intracellular signal transduction. The GCs are classified into two groups, particulate/membrane GCs (pGC) and soluble/cytosolic GCs (sGC). In relation to the endocrine system, pGCs include hormone receptors for natriuretic peptides (GC-A and GC-B) and guanylin peptides (GC-C), while sGC is a receptor for nitric oxide and carbon monoxide. Comparing the functions of pGCs in eukaryotes, it is apparent that pGCs perceive various environmental factors such as light, temperature, and various external chemical signals in addition to endocrine hormones, and transmit the information into the cell using the intracellular signaling cascade initiated by cGMP, e.g., cGMP-dependent protein kinases, cGMP-sensitive cyclic nucleotide-gated ion channels and cGMP-regulated phosphodiesterases. Among vertebrate pGCs, GC-E and GC-F are localized on retinal epithelia and are involved in modifying signal transduction from the photoreceptor, rhodopsin. GC-D and GC-G are localized in olfactory epithelia and serve as sensors at the extracellular domain for external chemical signals such as odorants and pheromones. GC-G also responds to guanylin peptides in the urine, which alters sensitivity to other chemicals. In addition, guanylin peptides that are secreted into the intestinal lumen, a pseudo-external environment, act on the GC-C on the apical membrane for regulation of epithelial transport. In this context, GC-C and GC-G appear to be in transition from exocrine pheromone receptor to endocrine hormone receptor. The pGCs also exist in various deuterostome and protostome invertebrates, and act as receptors for environmental, exocrine and endocrine factors including hormones. Tracing the evolutionary history of pGCs, it appears that pGCs first appeared as a sensor for physicochemical signals in the environment, and then evolved to function as hormone receptors. In this review, the author proposes an evolutionary history of pGCs that highlights the emerging role of the GC/cGMP system for signal transduction in hormone action.
Collapse
Affiliation(s)
- Yoshio Takei
- Laboratory of Physiology, Department of Marine Bioscience, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba 277-8564, Japan.
| |
Collapse
|
44
|
Meng QT, Liu XY, Liu XT, Liu J, Munanairi A, Barry DM, Liu B, Jin H, Sun Y, Yang Q, Gao F, Wan L, Peng J, Jin JH, Shen KF, Kim R, Yin J, Tao A, Chen ZF. BNP facilitates NMB-encoded histaminergic itch via NPRC-NMBR crosstalk. eLife 2021; 10:71689. [PMID: 34919054 PMCID: PMC8789279 DOI: 10.7554/elife.71689] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 12/16/2021] [Indexed: 11/13/2022] Open
Abstract
Histamine-dependent and -independent itch is conveyed by parallel peripheral neural pathways that express gastrin-releasing peptide (GRP) and neuromedin B (NMB), respectively, to the spinal cord of mice. B-type natriuretic peptide (BNP) has been proposed to transmit both types of itch via its receptor NPRA encoded by Npr1. However, BNP also binds to its cognate receptor, NPRC encoded by Npr3 with equal potency. Moreover, natriuretic peptides (NP) signal through the Gi-couped inhibitory cGMP pathway that is supposed to inhibit neuronal activity, raising the question of how BNP may transmit itch information. Here, we report that Npr3 expression in laminae I-II of the dorsal horn partially overlaps with NMB receptor (NMBR) that transmits histaminergic itch via Gq-couped PLCβ-Ca2+ signaling pathway. Functional studies indicate that NPRC is required for itch evoked by histamine but not chloroquine (CQ), a nonhistaminergic pruritogen. Importantly, BNP significantly facilitates scratching behaviors mediated by NMB, but not GRP. Consistently, BNP evoked Ca2+ responses in NMBR/NPRC HEK 293 cells and NMBR/NPRC dorsal horn neurons. These results reveal a previously unknown mechanism by which BNP facilitates NMB-encoded itch through a novel NPRC-NMBR cross-signaling in mice. Our studies uncover distinct modes of action for neuropeptides in transmission and modulation of itch in mice. An itch is a common sensation that makes us want to scratch. Most short-term itches are caused by histamine, a chemical that is released by immune cells following an infection or in response to an allergic reaction. Chronic itching, on the other hand, is not usually triggered by histamine, and is typically the result of neurological or skin disorders, such as atopic dermatitis. The sensation of itching is generated by signals that travel from the skin to nerve cells in the spinal cord. Studies in mice have shown that the neuropeptides responsible for delivering these signals differ depending on whether or not the itch involves histamine: GRPs (short for gastrin-releasing proteins) convey histamine-independent itches, while NMBs (short for neuromedin B) convey histamine-dependent itches. It has been proposed that another neuropeptide called BNP (short for B-type natriuretic peptide) is able to transmit both types of itch signals to the spinal cord. But it remains unclear how this signaling molecule is able to do this. To investigate, Meng, Liu, Liu, Liu et al. carried out a combination of behavioral, molecular and pharmacological experiments in mice and nerve cells cultured in a laboratory. The experiments showed that BNP alone cannot transmit the sensation of itching, but it can boost itching signals that are triggered by histamine. It is widely believed that BNP activates a receptor protein called NPRA. However, Meng et al. found that the BNP actually binds to another protein which alters the function of the receptor activated by NMBs. These findings suggest that BNP modulates rather than initiates histamine-dependent itching by enhancing the interaction between NMBs and their receptor. Understanding how itch signals travel from the skin to neurons in the spinal cord is crucial for designing new treatments for chronic itching. The work by Meng et al. suggests that treatments targeting NPRA, which was thought to be a key itch receptor, may not be effective against chronic itching, and that other drug targets need to be explored.
Collapse
Affiliation(s)
- Qing-Tao Meng
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Xian-Yu Liu
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Xue-Ting Liu
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Juan Liu
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Admire Munanairi
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Devin M Barry
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Benlong Liu
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Hua Jin
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Yu Sun
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Qianyi Yang
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Fang Gao
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Li Wan
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Jiahang Peng
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Jin-Hua Jin
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Kai-Feng Shen
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Ray Kim
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Jun Yin
- Center for the Study of Itch and Sensory Disorders, Washington University in St. Louis, St Louis, United States
| | - Ailin Tao
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhou-Feng Chen
- Department of Anesthesiology, Washington University in St. Louis, St Louis, United States
| |
Collapse
|
45
|
Jansen HJ, Moghtadaei M, Rafferty SA, Rose RA. Loss of natriuretic peptide receptor C enhances sinoatrial node dysfunction in aging and frail mice. J Gerontol A Biol Sci Med Sci 2021; 77:902-908. [PMID: 34865023 DOI: 10.1093/gerona/glab357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Indexed: 11/14/2022] Open
Abstract
Heart rate is controlled by the sinoatrial node (SAN). SAN dysfunction is highly prevalent in aging; however, not all individuals age at the same rate. Rather, health status during aging is affected by frailty. Natriuretic peptides regulate SAN function in part by activating natriuretic peptide receptor C (NPR-C). The impacts of NPR-C on HR and SAN function in aging and as a function of frailty are unknown. Frailty was measured in aging wildtype (WT) and NPR-C knockout (NPR-C -/-) mice using a mouse clinical frailty index (FI). HR and SAN structure and function were investigated using intracardiac electrophysiology in anesthetized mice, high-resolution optical mapping in intact atrial preparations, histology and molecular biology. NPR-C -/- mice rapidly became frail leading to shortened lifespan. HR and SAN recovery time were increased in older vs. younger mice and this was exacerbated in NPR-C -/- mice; however, there was substantial variability among age groups and genotypes. HR and SAN recovery time were correlated with FI score and fell along a continuum regardless of age or genotype. Optical mapping demonstrates impairments in SAN function that were also strongly correlated with FI score. SAN fibrosis was increased in aged and NPR-C -/- mice and was graded by FI score. Loss of NPR-C results in accelerated aging due to a rapid decline in health status in association with impairments in HR and SAN function. Frailty assessment was effective and often better able to distinguish aging-dependent changes in SAN function in the setting of shorted lifespan due to loss of NPR-C.
Collapse
Affiliation(s)
- Hailey J Jansen
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, Calgary, Alberta, Canada
| | - Motahareh Moghtadaei
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, Calgary, Alberta, Canada
| | - Sara A Rafferty
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Robert A Rose
- Libin Cardiovascular Institute, Department of Cardiac Sciences, Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
46
|
Tegin G, Gao Y, Hamlyn JM, Clark BJ, El-Mallakh RS. Inhibition of endogenous ouabain by atrial natriuretic peptide is a guanylyl cyclase independent effect. PLoS One 2021; 16:e0260131. [PMID: 34793577 PMCID: PMC8601428 DOI: 10.1371/journal.pone.0260131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 11/03/2021] [Indexed: 12/15/2022] Open
Abstract
Background Endogenous ouabain (EO) and atrial natriuretic peptide (ANP) are important in regulation of sodium and fluid balance. There is indirect evidence that ANP may be involved in the regulation of endogenous cardenolides. Methods H295R are human adrenocortical cells known to release EO. Cells were treated with ANP at physiologic concentrations or vehicle (0.1% DMSO), with or without guanylyl cyclase inhibitor 1,2,4 oxadiazolo[4,3-a]quinoxalin-1-one (ODQ). Cyclic guanosine monophosphate (cGMP), the intracellular second messenger of ANP, was measured by a chemiluminescent immunoassay and EO was measured by radioimmunoassay of C18 extracted samples. Results EO secretion is inhibited by ANP treatment, with the most prolonged inhibition (90 min vs ≤ 60 min) occurring at physiologic ANP concentrations (50 pg/mL). Inhibition of guanylyl cyclase with ODQ, also reduces EO secretion. The inhibitory effects on EO release in response to cotreatment with ANP and ODQ appeared to be additive. Conclusions ANP inhibits basal EO secretion, and it is unlikely that this is mediated through ANP-A or ANP-B receptors (the most common natriuretic peptide receptors) or their cGMP second messenger; the underlying mechanisms involved are not revealed in the current studies. The role of ANP in the control of EO synthesis and secretion in vivo requires further investigation.
Collapse
Affiliation(s)
- Gulay Tegin
- Department of Psychiatry and Behavioral Sciences, University of Louisville, Louisville, Kentucky, United States of America
| | - Yonglin Gao
- Department of Psychiatry and Behavioral Sciences, University of Louisville, Louisville, Kentucky, United States of America
| | - John M. Hamlyn
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Mississippi, United States of America
| | - Barbara J. Clark
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky, United States of America
| | - Rif S. El-Mallakh
- Department of Psychiatry and Behavioral Sciences, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
47
|
Handa T, Mori KP, Ishii A, Ohno S, Kanai Y, Watanabe-Takano H, Yasoda A, Kuwabara T, Takahashi N, Mochizuki N, Mukoyama M, Yanagita M, Yokoi H. Osteocrin ameliorates adriamycin nephropathy via p38 mitogen-activated protein kinase inhibition. Sci Rep 2021; 11:21835. [PMID: 34750411 PMCID: PMC8575949 DOI: 10.1038/s41598-021-01095-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/21/2021] [Indexed: 11/09/2022] Open
Abstract
Natriuretic peptides exert multiple effects by binding to natriuretic peptide receptors (NPRs). Osteocrin (OSTN) binds with high affinity to NPR-C, a clearance receptor for natriuretic peptides, and inhibits degradation of natriuretic peptides and consequently enhances guanylyl cyclase-A (GC-A/NPR1) signaling. However, the roles of OSTN in the kidney have not been well clarified. Adriamycin (ADR) nephropathy in wild-type mice showed albuminuria, glomerular basement membrane changes, increased podocyte injuries, infiltration of macrophages, and p38 mitogen-activated protein kinase (MAPK) activation. All these phenotypes were improved in OSTN- transgenic (Tg) mice and NPR3 knockout (KO) mice, with no further improvement in OSTN-Tg/NPR3 KO double mutant mice, indicating that OSTN works through NPR3. On the contrary, OSTN KO mice increased urinary albumin levels, and pharmacological blockade of p38 MAPK in OSTN KO mice ameliorated ADR nephropathy. In vitro, combination treatment with ANP and OSTN, or FR167653, p38 MAPK inhibitor, reduced Ccl2 and Des mRNA expression in murine podocytes (MPC5). OSTN increased intracellular cyclic guanosine monophosphate (cGMP) in MPC5 through GC-A. We have elucidated that circulating OSTN improves ADR nephropathy by enhancing GC-A signaling and consequently suppressing p38 MAPK activation. These results suggest that OSTN could be a promising therapeutic agent for podocyte injury.
Collapse
Affiliation(s)
- Takaya Handa
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Keita P Mori
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan.,Department of Nephrology and Dialysis, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka, Japan.,TMK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Ishii
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Shoko Ohno
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Yugo Kanai
- Department of Diabetes Mellitus and Endocrinology, Osaka Red Cross Hospital, Osaka, Japan
| | - Haruko Watanabe-Takano
- Department of Cell Biology, National Cerebral and Cardiovascular Center, Research Institute, Suita, Japan
| | - Akihiro Yasoda
- Clinical Research Center, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Takashige Kuwabara
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Nobuyuki Takahashi
- Department of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Medicine, Sendai, Japan
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center, Research Institute, Suita, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan.,Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Hideki Yokoi
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan.
| |
Collapse
|
48
|
Gidlöf O. Toward a New Paradigm for Targeted Natriuretic Peptide Enhancement in Heart Failure. Front Physiol 2021; 12:650124. [PMID: 34721050 PMCID: PMC8548580 DOI: 10.3389/fphys.2021.650124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 09/21/2021] [Indexed: 12/11/2022] Open
Abstract
The natriuretic peptide system (NPS) plays a fundamental role in maintaining cardiorenal homeostasis, and its potent filling pressure-regulated diuretic and vasodilatory effects constitute a beneficial compensatory mechanism in heart failure (HF). Leveraging the NPS for therapeutic benefit in HF has been the subject of intense investigation during the last three decades and has ultimately reached widespread clinical use in the form of angiotensin receptor-neprilysin inhibition (ARNi). NPS enhancement via ARNi confers beneficial effects on mortality and hospitalization in HF, but inhibition of neprilysin leads to the accumulation of a number of other vasoactive peptides in the circulation, often resulting in hypotension and raising potential concerns over long-term adverse effects. Moreover, ARNi is less effective in the large group of HF patients with preserved ejection fraction. Alternative approaches for therapeutic augmentation of the NPS with increased specificity and efficacy are therefore warranted, and are now becoming feasible particularly with recent development of RNA therapeutics. In this review, the current state-of-the-art in terms of experimental and clinical strategies for NPS augmentation and their implementation will be reviewed and discussed.
Collapse
Affiliation(s)
- Olof Gidlöf
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
49
|
Liu LP, Gholam MF, Elshikha AS, Kawakibi T, Elmoujahid N, Moussa HH, Song S, Alli AA. Transgenic Mice Overexpressing Human Alpha-1 Antitrypsin Exhibit Low Blood Pressure and Altered Epithelial Transport Mechanisms in the Inactive and Active Cycles. Front Physiol 2021; 12:710313. [PMID: 34630137 PMCID: PMC8493122 DOI: 10.3389/fphys.2021.710313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/20/2021] [Indexed: 11/13/2022] Open
Abstract
Human alpha-1 antitrypsin (hAAT) is a versatile protease inhibitor, but little is known about its targets in the aldosterone-sensitive distal nephron and its role in electrolyte balance and blood pressure control. We analyzed urinary electrolytes, osmolality, and blood pressure from hAAT transgenic (hAAT-Tg) mice and C57B/6 wild-type control mice maintained on either a normal salt or high salt diet. Urinary sodium, potassium, and chloride concentrations as well as urinary osmolality were lower in hAAT-Tg mice maintained on a high salt diet during both the active and inactive cycles. hAAT-Tg mice showed a lower systolic blood pressure compared to C57B6 mice when maintained on a normal salt diet but this was not observed when they were maintained on a high salt diet. Cathepsin B protein activity was less in hAAT-Tg mice compared to wild-type controls. Protein expression of the alpha subunit of the sodium epithelial channel (ENaC) alpha was also reduced in the hAAT-Tg mice. Natriuretic peptide receptor C (NPRC) protein expression in membrane fractions of the kidney cortex was reduced while circulating levels of atrial natriuretic peptide (ANP) were greater in hAAT-Tg mice compared to wild-type controls. This study characterizes the electrolyte and blood pressure phenotype of hAAT-Tg mice during the inactive and active cycles and investigates the mechanism by which ENaC activation is inhibited in part by a mechanism involving decreased cathepsin B activity and increased ANP levels in the systemic circulation.
Collapse
Affiliation(s)
- Lauren P Liu
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Mohammed F Gholam
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Ahmed Samir Elshikha
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Tamim Kawakibi
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Nasseem Elmoujahid
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Hassan H Moussa
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Sihong Song
- Department of Pharmaceutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Abdel A Alli
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States.,Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
50
|
Jansen HJ, Moghtadaei M, Rafferty SA, Rose RA. Atrial Fibrillation in Aging and Frail Mice: Modulation by Natriuretic Peptide Receptor C. Circ Arrhythm Electrophysiol 2021; 14:e010077. [PMID: 34490788 DOI: 10.1161/circep.121.010077] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hailey J Jansen
- Department of Cardiac Sciences, Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada (H.J.J., M.M., R.A.R.)
| | - Motahareh Moghtadaei
- Department of Cardiac Sciences, Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada (H.J.J., M.M., R.A.R.)
| | - Sara A Rafferty
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada (S.A.R.)
| | - Robert A Rose
- Department of Cardiac Sciences, Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada (H.J.J., M.M., R.A.R.)
| |
Collapse
|