1
|
Shero JA, Lindholm ME, Sandri M, Stanford KI. Skeletal Muscle as a Mediator of Interorgan Crosstalk During Exercise: Implications for Aging and Obesity. Circ Res 2025; 136:1407-1432. [PMID: 40403102 PMCID: PMC12101524 DOI: 10.1161/circresaha.124.325614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 05/24/2025]
Abstract
Physical exercise is critical for preventing and managing chronic conditions, such as cardiovascular disease, type 2 diabetes, hypertension, and sarcopenia. Regular physical activity significantly reduces cardiovascular and all-cause mortality. Exercise also enhances metabolic health by promoting muscle growth, mitochondrial biogenesis, and improved nutrient storage while preventing age-related muscle dysfunction. Key metabolic benefits include increased glucose uptake, enhanced fat oxidation, and the release of exercise-induced molecules called myokines, which mediate interorgan communication and improve overall metabolic function. These myokines and other exercise-induced signaling molecules hold promise as therapeutic targets for aging and obesity-related conditions.
Collapse
Affiliation(s)
- Julia A. Shero
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Maléne E. Lindholm
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, California, United States
| | - Marco Sandri
- Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy
| | - Kristin I. Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| |
Collapse
|
2
|
Saw EL, Werner LD, Cooper HL, Pimental DR, Zamani P, Chirinos JA, Valero-Muñoz M, Sam F. Musclin Counteracts Skeletal Muscle Dysfunction and Exercise Intolerance in Heart Failure With Preserved Ejection Fraction. Circ Heart Fail 2025:e012350. [PMID: 40358602 DOI: 10.1161/circheartfailure.124.012350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 02/27/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Exercise intolerance is a hallmark of heart failure with preserved ejection fraction (HFpEF) and is characterized by skeletal muscle (SkM) dysfunction with impaired oxidative capacity. To maintain oxidative capacity, the SkM secretes myokines such as musclin, which has been shown to potentiate NP (natriuretic peptide) signaling and induce PGC-1α (peroxisome proliferator-activated receptor-γ coactivator-1 alpha) signaling. We sought to investigate the role of musclin in SkM dysfunction in HFpEF. For this study, we selected the oxidative-predominant SkM soleus in HFpEF mice and vastus lateralis from patients with HFpEF. METHODS Using the SAUNA model, mice underwent HFpEF induction by uninephrectomy, d-aldosterone infusion, and 1% sodium chloride drinking water for 4 weeks. Exogenous musclin was given to HFpEF mice every 2 days during the last 2 weeks of HFpEF induction. Molecular analyses were conducted on blood samples and SkM from HFpEF mice and patients with HFpEF. RESULTS In HFpEF mice and patients with HFpEF, increased musclin expression was accompanied by decreased cyclic guanosine monophosphate levels and PGC-1α expression in SkM, suggesting impaired NP signaling. Exogenous administration of musclin in mice with HFpEF demonstrated augmented circulating musclin levels and potentiated NP signaling in SkM as shown by increased PKG1 (protein kinase G1) activity and PGC-1α expression. This was associated with a transition from type-2A to type-1 fiber (type-1 has more endurance) and increased succinate dehydrogenase activity, hindlimb blood flow, and capillary density in the soleus muscle. Exogenous musclin also mitigated cardiac hypertrophy without affecting blood pressure or diastolic function. Most importantly, HFpEF mice treated with musclin demonstrated improved functional and exercise capacity. CONCLUSIONS Musclin mediates beneficial effects in SkM and heart with improved exercise capacity likely by improving oxidative capacity in SkM. Future studies are warranted to address the therapeutic efficacy of exogenous musclin in humans with HFpEF.
Collapse
Affiliation(s)
- Eng Leng Saw
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, MA (E.L.S., L.D.W., H.L.C., D.R.P., M.V.-M., F.S.)
| | - Louis Dominic Werner
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, MA (E.L.S., L.D.W., H.L.C., D.R.P., M.V.-M., F.S.)
| | - Hannah L Cooper
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, MA (E.L.S., L.D.W., H.L.C., D.R.P., M.V.-M., F.S.)
| | - David R Pimental
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, MA (E.L.S., L.D.W., H.L.C., D.R.P., M.V.-M., F.S.)
| | - Payman Zamani
- Division of Cardiovascular Medicine, Penn Cardiovascular Institute, Hospital of the University of Pennsylvania, Philadelphia (P.Z., J.A.C.)
| | - Julio A Chirinos
- Division of Cardiovascular Medicine, Penn Cardiovascular Institute, Hospital of the University of Pennsylvania, Philadelphia (P.Z., J.A.C.)
| | - María Valero-Muñoz
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, MA (E.L.S., L.D.W., H.L.C., D.R.P., M.V.-M., F.S.)
| | - Flora Sam
- Department of Medicine, Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, MA (E.L.S., L.D.W., H.L.C., D.R.P., M.V.-M., F.S.)
| |
Collapse
|
3
|
Dong N, Du M, Wu Q. Molecular insights into the corin function at the uteroplacental interface. Placenta 2025:S0143-4004(25)00159-6. [PMID: 40360315 DOI: 10.1016/j.placenta.2025.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/24/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
In pregnancy, cell-cell interactions and tissue remodeling are important physiological processes at the uteroplacental interface. To date, molecular mechanisms governing cell activities at the uteroplacental interface are not fully understood. Corin is a proteolytic enzyme responsible for activating atrial natriuretic peptide (ANP), a multifunctional hormone essential for cardiovascular and metabolic homeostasis. Upon progesterone stimulation, corin expression is induced in the uterus via a specific set of transcription factors. Uterine corin activates ANP to enhance decidualization and cell-cell interactions within the vasculature, leading to sequential vascular smooth muscle and endothelial cell death in spiral arteries. These events are crucial for uterine vascular remodeling and trophoblast invasion. Corin also functions in the decidua to regulate macrophage distribution and function in response to placental ischemia. In mice, Corin knockout impairs endometrial decidualization, vascular remodeling, and macrophage function at the uteroplacental interface, causing a preeclampsia (PE)-like phenotype. In humans, deleterious variants and impaired epigenetic modifications in the CORIN gene have been reported in women with PE, indicating that corin deficiency may be a contributing factor in the pathogenesis of PE. In this review, we describe the corin function at the uteroplacental interface and underlying molecular mechanisms. We also discuss potential implications of corin deficiency in pregnancy-associated diseases.
Collapse
Affiliation(s)
- Ningzheng Dong
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China; Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Meirong Du
- Laboratory of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University Shanghai Medical College, China
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China.
| |
Collapse
|
4
|
Nicoli CD, Long DL, Plante TB, Judd SE, McClure LA, Carson AP, Cushman M. N-terminal Pro-B-Type Natriuretic Peptide and Risk for Diabetes Mellitus and Metabolic Syndrome. J Clin Endocrinol Metab 2025; 110:e1185-e1193. [PMID: 38703102 PMCID: PMC11913105 DOI: 10.1210/clinem/dgae301] [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: 02/05/2024] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024]
Abstract
CONTEXT Natriuretic peptide concentrations are inversely associated with risk of diabetes mellitus and may be protective from metabolic dysfunction. OBJECTIVE We studied associations of N-terminal pro-B-type natriuretic peptide (NT-proBNP) with incident diabetes, metabolic syndrome (MetS), and MetS components. METHODS A total of 2899 participants with baseline (2003-2007) and follow-up (2013-2016) examinations and baseline NT-proBNP measurement in the REasons for Geographic And Racial Differences in Stroke study. Logistic regression models were fitted to incident MetS, MetS components, and diabetes; covariates included demographics, risk and laboratory factors. Incident diabetes was defined as fasting glucose ≥126 mg/dL, random glucose ≥200 mg/dL, or use of insulin or hypoglycemic drugs at follow-up but not baseline. Incident MetS was defined as participants with ≥3 harmonized criteria at follow-up and <3 at baseline. RESULTS A total of 310 participants (2364 at risk) developed diabetes and 361 (2059 at risk) developed MetS over a mean 9.4 years of follow-up. NT-proBNP was inversely associated with odds of incident diabetes (fully adjusted OR per SD higher log NT-proBNP 0.80, 95% CI 0.69-0.93) and MetS in the highest vs lowest quartile only (fully adjusted OR 0.59, 95% CI 0.37-0.92); the linear association with incident MetS was not statistically significant. NT-proBNP was inversely associated with incident dysglycemia in all models (fully adjusted OR per SD log NT-proBNP 0.65, 95% CI 0.53-0.79), but not with other MetS components. Effect modification by sex, race, age, or body mass index was not observed. CONCLUSION NT-proBNP was inversely associated with odds of diabetes, MetS, and the MetS dysglycemia component. The metabolic implications of B-type natriuretic peptides appear important for glycemic homeostasis.
Collapse
Affiliation(s)
- Charles D Nicoli
- Department of Medicine, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
| | - D Leann Long
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Timothy B Plante
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT 05401, USA
| | - Suzanne E Judd
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Leslie A McClure
- Department of Epidemiology & Biostatistics, Dornsife School of Public Health, Drexel University, Philadelphia, PA 19104, USA
| | - April P Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Mary Cushman
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT 05401, USA
- Department of Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, Burlington, VT 05401, USA
| |
Collapse
|
5
|
Katz DH, Lindholm ME, Ashley EA. Charting the Molecular Terrain of Exercise: Energetics, Exerkines, and the Future of Multiomic Mapping. Physiology (Bethesda) 2025; 40:0. [PMID: 39136551 DOI: 10.1152/physiol.00024.2024] [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: 05/07/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 11/21/2024] Open
Abstract
Physical activity plays a fundamental role in human health and disease. Exercise has been shown to improve a wide variety of disease states, and the scientific community is committed to understanding the precise molecular mechanisms that underlie the exquisite benefits. This review provides an overview of molecular responses to acute exercise and chronic training, particularly energy mobilization and generation, structural adaptation, inflammation, and immune regulation. Furthermore, it offers a detailed discussion of known molecular signals and systemic regulators activated during various forms of exercise and their role in orchestrating health benefits. Critically, the increasing use of multiomic technologies is explored with an emphasis on how multiomic and multitissue studies contribute to a more profound understanding of exercise biology. These data inform anticipated future advancement in the field and highlight the prospect of integrating exercise with pharmacology for personalized disease prevention and treatment.
Collapse
Affiliation(s)
- Daniel H Katz
- Division of Cardiovascular MedicineStanford University School of Medicine, Stanford, California, United States
| | - Maléne E Lindholm
- Division of Cardiovascular MedicineStanford University School of Medicine, Stanford, California, United States
| | - Euan A Ashley
- Division of Cardiovascular MedicineStanford University School of Medicine, Stanford, California, United States
| |
Collapse
|
6
|
Prickett TCR, Lewis LK, Pearson JF, Espiner EA. Metabolism of Natriuretic peptides and impact on insulin resistance and fat mass in healthy subjects. Clin Biochem 2025; 136:110893. [PMID: 39914781 DOI: 10.1016/j.clinbiochem.2025.110893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/26/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Natriuretic peptides (NP) have important roles in regulating fat balance and metabolic health. Reduced concentrations of ANP and BNP in plasma are associated with increased insulin resistance in obesity. Whether this is due to increased clearance or reduced bioactivity of immunoreactive NP forms is unclear. DESIGN AND METHOD These questions were addressed in a community study of mildly obese subjects at middle age. The ratio of amino-terminal (NT) pro-NP to bioactive C-terminal NP was used as a putative index of the clearance of bioactive forms. RESULTS Lower ratios of amino-terminal pro-NP to bioactive C-terminal NP were associated with increased insulin resistance. In linear regression models, NT-proANP and NT-proBNP outperformed ANP and BNP in predicting insulin resistance. Pro-NP glycosylation, which can impair NP and NT-proNP production in obesity, does not account for the diminished impact of ANP or BNP. Plasma concentrations of osteocrin, which competes for the NP clearance receptor (NPR-C) and potentially enhances NP bioactivity, was not associated with NPs, but did positively predict insulin resistance in females. CONCLUSIONS We find no evidence that increased clearance/degradation of NPs contributes to insulin resistance. Among the nine NP variants assessed, only NT-proANP and NT-proBNP independently predicted insulin resistance in both sexes. The impact of CNP on fat mass or insulin resistance was minor but significant in females. Lower concentrations of immunoreactive plasma ANP and BNP remains unexplained and requires closer study.
Collapse
Affiliation(s)
| | - Lynley K Lewis
- Departments of Medicine, University of Otago, Christchurch, New Zealand
| | - John F Pearson
- Departments of Medicine, University of Otago, Christchurch, New Zealand; Biostatistics and Computational Biology Unit, University of Otago, Christchurch, New Zealand
| | - Eric A Espiner
- Departments of Medicine, University of Otago, Christchurch, New Zealand
| |
Collapse
|
7
|
Huang PF, Wang QY, Chen RB, Wang YD, Wang YY, Liu JH, Xiao XH, Liao ZZ. A New Strategy for Obesity Treatment: Revealing the Frontiers of Anti-obesity Medications. Curr Mol Med 2025; 25:13-26. [PMID: 38289639 DOI: 10.2174/0115665240270426231123155924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 02/19/2025]
Abstract
Obesity dramatically increases the risk of type 2 diabetes, fatty liver, hypertension, cardiovascular disease, and cancer, causing both declines in quality of life and life expectancy, which is a serious worldwide epidemic. At present, more and more patients with obesity are choosing drug therapy. However, given the high failure rate, high cost, and long design and testing process for discovering and developing new anti-obesity drugs, drug repurposing could be an innovative method and opportunity to broaden and improve pharmacological tools in this context. Because different diseases share molecular pathways and targets in the cells, anti-obesity drugs discovered in other fields are a viable option for treating obesity. Recently, some drugs initially developed for other diseases, such as treating diabetes, tumors, depression, alcoholism, erectile dysfunction, and Parkinson's disease, have been found to exert potential anti-obesity effects, which provides another treatment prospect. In this review, we will discuss the potential benefits and barriers associated with these drugs being used as obesity medications by focusing on their mechanisms of action when treating obesity. This could be a viable strategy for treating obesity as a significant advance in human health.
Collapse
Affiliation(s)
- Pan-Feng Huang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qi-Yu Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Rong-Bin Chen
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Ya-Di Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yuan-Yuan Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jiang-Hua Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xin-Hua Xiao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhe-Zhen Liao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| |
Collapse
|
8
|
Zheng Z, Liang J, Gao Y, Hua M, Zhang S, Liu M, Fang Z. Serum N-Terminal Pro-B-Type Natriuretic Peptide Is Associated With Insulin Resistance in Chinese: Danyang Study. J Clin Hypertens (Greenwich) 2024; 26:1256-1263. [PMID: 39311705 PMCID: PMC11555537 DOI: 10.1111/jch.14906] [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: 07/29/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 11/13/2024]
Abstract
The association of serum N-terminal pro-B-type natriuretic peptide (NT-proBNP) with insulin resistance (IR), as measured by homeostasis model assessment of insulin resistance (HOMA-IR), in the general population is unclear. Our study aimed to characterize its relationship in a large community-based population. Subjects were recruited from the Danyang city between 2017 and 2019. Serum NT-proBNP was measured using an enhanced chemiluminescence immunoassay. IR was defined by a HOMA-IR in the highest sex-specific quartile. Categorical and continuous analyses were performed with sex-specific NT-proBNP tertiles and naturally logarithmically transformed NT-proBNP (lnNTproBNP), respectively. The 2945 participants (mean age 52.8 years) included 1728 (58.7%) women, 1167 (39.6%) hypertensive patients, 269 (9.1%) diabetic patients, and 736 (25.0%) patients with IR. In simple and multivariate-adjusted regression analyses, serum lnNTproBNP were both negatively associated with HOMA-IR (β = -0.19 to -0.25; p < 0.0001). Similar results were also obtained in multiple subgroup analyses. In multiple logistic regression analyses, elevated serum NT-proBNP was associated with lower risks of IR (odds ratios: 0.68 and 0.39; 95% confidence intervals: 0.61-0.74 and 0.30-0.50 for lnNTproBNP and top vs. bottom tertiles, respectively; p < 0.0001). In conclusion, increased serum NT-proBNP level was strongly associated with a lower risk of IR in Chinese.
Collapse
Affiliation(s)
- Ziwen Zheng
- Department of CardiologyAffiliated Hospital of Nanjing University of Chinese MedicineJiangsu Province Hospital of Chinese MedicineNanjingJiangsuChina
| | - Junya Liang
- Institute of HypertensionAffiliated Hospital of Nanjing University of Chinese MedicineJiangsu Province Hospital of Chinese MedicineNanjingJiangsuChina
| | - Yun Gao
- Institute of HypertensionAffiliated Hospital of Nanjing University of Chinese MedicineJiangsu Province Hospital of Chinese MedicineNanjingJiangsuChina
| | - Mulian Hua
- Institute of HypertensionAffiliated Hospital of Nanjing University of Chinese MedicineJiangsu Province Hospital of Chinese MedicineNanjingJiangsuChina
| | - Siqi Zhang
- Institute of HypertensionAffiliated Hospital of Nanjing University of Chinese MedicineJiangsu Province Hospital of Chinese MedicineNanjingJiangsuChina
| | - Ming Liu
- Department of CardiologyAffiliated Hospital of Nanjing University of Chinese MedicineJiangsu Province Hospital of Chinese MedicineNanjingJiangsuChina
- Institute of HypertensionAffiliated Hospital of Nanjing University of Chinese MedicineJiangsu Province Hospital of Chinese MedicineNanjingJiangsuChina
| | - Zhuyuan Fang
- Department of CardiologyAffiliated Hospital of Nanjing University of Chinese MedicineJiangsu Province Hospital of Chinese MedicineNanjingJiangsuChina
- Institute of HypertensionAffiliated Hospital of Nanjing University of Chinese MedicineJiangsu Province Hospital of Chinese MedicineNanjingJiangsuChina
| |
Collapse
|
9
|
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
|
10
|
Okuyama T, Nagoshi T, Hiraki N, Tanaka TD, Oi Y, Kimura H, Kashiwagi Y, Ogawa K, Minai K, Ogawa T, Kawai M, Yoshimura M. Blunted increase in plasma BNP during acute coronary syndrome attacks in obese patients. IJC HEART & VASCULATURE 2024; 54:101508. [PMID: 39314921 PMCID: PMC11417597 DOI: 10.1016/j.ijcha.2024.101508] [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: 06/24/2024] [Revised: 08/14/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024]
Abstract
Background Unexpectedly low natriuretic peptide (NP) levels in proportion to heart failure severity are often observed in obese individuals. However, the magnitude of NP elevation in response to acute cardiac stress in obesity has not yet been extensively studied. This study aimed to determine the impact of obesity on the increase in plasma NP in response to cardiac hemodynamic stress during acute coronary syndrome (ACS) attacks. Methods and Results The study population included 557 consecutive patients with ACS for whom data were collected during emergency cardiac catheterization. To determine the possible impact of body mass index (BMI) on the relationship between left ventricular ejection fraction (LVEF) and plasma B-type NP (BNP) levels, the study population was divided into two groups (Group 1: BMI <25, Group 2: BMI ≥25 [kg/m2]). Both BMI and LVEF were significantly and negatively correlated with BNP. Although a significant negative correlation between LVEF and BNP was observed in both groups, the regression line of Group 2 was significantly less steep than that of Group 1. Accordingly, BNP/LVEF ratio in Group 2, which indicates the extent of BNP increase in response to LVEF change, was significantly lower than that in Group 1. Conclusions Blunted increase in plasma BNP in response to cardiac hemodynamic stress during ACS attacks was observed in obese individuals. In addition to the relatively low plasma BNP levels at baseline in obese individuals, the blunted response of BNP elevation to ACS attacks may have important pathophysiological implications for hemodynamic regulation and myocardial energy metabolism.
Collapse
Affiliation(s)
| | | | - Nana Hiraki
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Toshikazu D. Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Yuhei Oi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Haruka Kimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Yusuke Kashiwagi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Kazuo Ogawa
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Kosuke Minai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Takayuki Ogawa
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| |
Collapse
|
11
|
Potter LR. Phosphorylation-Dependent Regulation of Guanylyl Cyclase (GC)-A and Other Membrane GC Receptors. Endocr Rev 2024; 45:755-771. [PMID: 38713083 PMCID: PMC11405504 DOI: 10.1210/endrev/bnae015] [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: 12/15/2023] [Revised: 04/07/2024] [Accepted: 05/01/2024] [Indexed: 05/08/2024]
Abstract
Receptor guanylyl cyclases (GCs) are single membrane spanning, multidomain enzymes, that synthesize cGMP in response to natriuretic peptides or other ligands. They are evolutionarily conserved from sea urchins to humans and regulate diverse physiologies. Most family members are phosphorylated on 4 to 7 conserved serines or threonines at the beginning of their kinase homology domains. This review describes studies that demonstrate that phosphorylation and dephosphorylation are required for activation and inactivation of these enzymes, respectively. Phosphorylation sites in GC-A, GC-B, GC-E, and sea urchin receptors are discussed, as are mutant receptors that mimic the dephosphorylated inactive or phosphorylated active forms of GC-A and GC-B, respectively. A salt bridge model is described that explains why phosphorylation is required for enzyme activation. Potential kinases, phosphatases, and ATP regulation of GC receptors are also discussed. Critically, knock-in mice with glutamate substitutions for receptor phosphorylation sites are described. The inability of opposing signaling pathways to inhibit cGMP synthesis in mice where GC-A or GC-B cannot be dephosphorylated demonstrates the necessity of receptor dephosphorylation in vivo. Cardiac hypertrophy, oocyte meiosis, long-bone growth/achondroplasia, and bone density are regulated by GC phosphorylation, but additional processes are likely to be identified in the future.
Collapse
Affiliation(s)
- Lincoln R Potter
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
12
|
Cherezova A, Sudarikova A, Vasileva V, Iurchenko R, Nikiforova A, Spires DR, Zamaro AS, Jones AC, Schibalski RS, Dong Z, Palygin O, Stadler K, Ilatovskaya DV. The effects of the atrial natriuretic peptide deficiency on renal cortical mitochondrial bioenergetics in the Dahl SS rat. FASEB J 2024; 38:e23891. [PMID: 39150822 PMCID: PMC11335316 DOI: 10.1096/fj.202400672rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/18/2024]
Abstract
Atrial Natriuretic Peptide (ANP) plays an important role in blood pressure regulation. Low levels of ANP correlate with the development of salt-sensitive hypertension (SS-HTN). Our previous studies indicated that ANP deficiency exacerbated renal function decline in SS-HTN. In the heart and fat tissue, ANP was reported to affect lipid peroxidation and mitochondrial bioenergetics but the effects of ANP on mitochondrial function in the kidney are unexplored. We hypothesized that ANP deficiency in SS-HTN causes renal bioenergetic shift, leading to disruption of mitochondrial network and oxidative stress. To address the hypothesis, we placed Dahl SS wild-type (SSWT) and ANP knockout (SSNPPA-/-) rats on 4% NaCl high salt (HS) diet to induce HTN or maintained them on 0.4% NaCl normal salt (NS) diet and assessed mitochondrial bioenergetics and dynamics using spectrofluorimetry, Seahorse assay, electron paramagnetic resonance (EPR) spectroscopy, Western blotting, electron microscopy, PCR and cytokine assays. We report that under high salt conditions, associated with hypertension and renal damage, the SSNPPA-/- rats exhibit a decrease in mitochondrial membrane potential and elevation in mitochondrial ROS levels compared to SSWT. The redox shift is also evident by the presence of more pronounced medullar lipid peroxidation in the SSNPPA-/- strain. We also revealed fragmented, more damaged mitochondria in the SSNPPA-/- rats, accompanied by increased turnover and biogenesis. Overall, our data indicate that ANP deficiency causes disruptions in mitochondrial bioenergetics and dynamics which likely contributes to aggravation of the renal damage and hypertension in the Dahl SS rat; the major pathological effects are evident in the groups subjected to a combined salt and ANP deficiency-induced mitochondrial stress.
Collapse
Affiliation(s)
- Alena Cherezova
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, 30912, USA
| | - Anastasia Sudarikova
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, 30912, USA
| | - Valeria Vasileva
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, 30912, USA
| | - Regina Iurchenko
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, 29425, USA
| | - Anna Nikiforova
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, 29425, USA
| | - Denisha R. Spires
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, 30912, USA
| | - Aleksandra S. Zamaro
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, 30912, USA
| | - Adam C. Jones
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, 30912, USA
| | - Ryan S. Schibalski
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, 30912, USA
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, 30912, USA
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, 29425, USA
| | | | - Daria V. Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, 30912, USA
| |
Collapse
|
13
|
Bachmann KN, Ceddia RP, Gupta DK, Collins S, Wang TJ. Human adipose tissue expression of uncoupling protein 1 in response to intravenous administration of B-type natriuretic peptide hormone: Results from a randomized controlled crossover study. Diabetes Obes Metab 2024; 26:3458-3461. [PMID: 38686535 PMCID: PMC12036528 DOI: 10.1111/dom.15628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/02/2024] [Accepted: 04/14/2024] [Indexed: 05/02/2024]
Affiliation(s)
- Katherine N. Bachmann
- Office of Research and Development, Clinical Sciences Research and Development (CSR&D), United States Department of Veterans Affairs, Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt Translational and Clinical Cardiovascular Research Center, Nashville, Tennessee, USA
| | - Ryan P. Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Deepak K. Gupta
- Department of Medicine, Vanderbilt Translational and Clinical Cardiovascular Research Center, Nashville, Tennessee, USA
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Thomas J. Wang
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
14
|
Kashiwagi Y, Nagoshi T, Tanaka Y, Oi Y, Kimura H, Ogawa K, Kawai M, Yoshimura M. Effects of angiotensin receptor-neprilysin inhibitor on ketone body metabolism in pre-heart failure/heart failure patients. Sci Rep 2024; 14:16493. [PMID: 39020009 PMCID: PMC11255280 DOI: 10.1038/s41598-024-67524-6] [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: 03/02/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024] Open
Abstract
Recently, a mild elevation of the blood ketone levels was found to exert multifaceted cardioprotective effects. To investigate the effect of angiotensin receptor neprilysin inhibitors (ARNIs) on the blood ketone body levels, 46 stable pre-heart failure (HF)/HF patients were studied, including 23 who switched from angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs) to ARNIs (ARNI group) and 23 who continued treatment with ACE inhibitors or ARBs (control group). At baseline, there were no significant differences in the total ketone body (TKB) levels between the two groups. Three months later, the TKB levels in the ARNI group were higher than the baseline values (baseline to 3 months: 71 [51, 122] to 92 [61, 270] μmol/L, P < 0.01). In the control group, no significant change was observed between the baseline and 3 months later. A multiple regression analysis demonstrated that the initiation of ARNI and an increase in the blood non-esterified fatty acid (NEFA) levels at 3 months increased the percentage changes in the TKB levels from baseline to 3 months (%ΔTKB level) (initiation of ARNI: P = 0.017, NEFA level at 3 months: P < 0.001). These results indicate that ARNI administration induces a mild elevation of the blood TKB levels in pre-HF/HF patients.
Collapse
Affiliation(s)
- Yusuke Kashiwagi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan.
| | - Tomohisa Nagoshi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yoshiro Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yuhei Oi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Haruka Kimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Kazuo Ogawa
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| |
Collapse
|
15
|
Zhang Z, Jia Z. Pre-IVM with C-type natriuretic peptide promotes mitochondrial biogenesis of bovine oocytes via activation of CREB. Sci Rep 2024; 14:16260. [PMID: 39009622 PMCID: PMC11250819 DOI: 10.1038/s41598-024-67094-7] [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: 05/27/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
The aim of this study was to evaluate the effects of C-type natriuretic peptide (CNP) treatment prior to in vitro maturation (IVM) on mitochondria biogenesis in bovine oocyte matured in vitro and explore the related causes. The results showed that treatment with CNP before IVM significantly improved mitochondrial content, elevated the expression of genes related to mitochondria biogenesis, and increased the protein levels of phosphorylation of cAMP-response element binding protein (p-CREB) in bovine oocytes following IVM. However, further studies revealed that treatment with CNP before IVM could not increased the protein levels of p-CREB in bovine oocytes when natriuretic peptide receptor 2 activities was inhibited using the relative specific inhibitor Gö6976. In addition, treatment with CNP before IVM could not improved mitochondrial content or elevated the expression of genes related to mitochondria biogenesis in bovine oocytes when CREB activities was abolished using the specific inhibitor 666-15. In summary, these results provide evidence that treatment of bovine oocytes with CNP before IVM promotes mitochondrial biogenesis in vitro, possibly by activating CREB.
Collapse
Affiliation(s)
- Zehua Zhang
- College of Animal Science and Technology, Inner Mongolia Minzu University, 536 West Huolinhe Street, Tongliao, 028000, Inner Mongolia, People's Republic of China
| | - Zhenwei Jia
- College of Animal Science and Technology, Inner Mongolia Minzu University, 536 West Huolinhe Street, Tongliao, 028000, Inner Mongolia, People's Republic of China.
| |
Collapse
|
16
|
Okamoto C, Tsukamoto O, Hasegawa T, Matsuoka K, Amaki M, Kanzaki H, Izumi C, Takashima S, Ito S, Kitakaze M. Relative B-Type Natriuretic Peptide Deficiency May Exist in Diastolic Dysfunction in Subclinical Population. Circ Rep 2024; 6:151-160. [PMID: 38736848 PMCID: PMC11081706 DOI: 10.1253/circrep.cr-24-0026] [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: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 05/14/2024] Open
Abstract
Background: Heart failure patients are deficient in B-type natriuretic peptide (BNP) but the significance of subclinical BNP deficiency is unclear. Methods and Results: A total of 1,398 subjects without cardiovascular disease, with left ventricular ejection fraction (LVEF) ≥50% and BNP level <100 pg/mL, were selected from a 2005-2008 health checkup in Arita-cho, Japan, and divided into 2 groups: with and without LV diastolic dysfunction (DD+ or DD-). We performed propensity score matching on non-cardiac factors affecting BNP levels and analyzed 470 subjects in each group (372/940 men; median age, 66 years). The DD(+) group showed higher lateral E/e', an index of estimated left ventricular filling pressure, and greater prevalence of concentric hypertrophy (CH) despite similar BNP levels, suggesting a relative deficiency of BNP in DD(+) compared with DD(-). Multivariable logistic regression analysis revealed an increase in BNP correlated with decreased odds of CH (adjusted odds ratio [aOR] 0.663, 95% confidence interval (CI) 0.484-0.909, P=0.011), whereas an increase in lateral E/e' was associated with increased odds of CH (aOR, 2.881; 95% CI, 1.390-5.973; P=0.004). Furthermore, CH in combination with diastolic dysfunction independently predicted major adverse cardiovascular events (hazard ratio 3.272, 95% CI 1.215-8.809; P=0.019). Conclusions: Relative BNP deficiency was associated with CH, which had a poor prognosis in patients with diastolic dysfunction.
Collapse
Affiliation(s)
- Chisato Okamoto
- Department of Cardiovascular Medicine, Hanwa Memorial HospitalOsakaJapan
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier BiosciencesSuitaJapan
- The Osaka Medical Research Foundation for Intractable DiseasesOsakaJapan
| | - Osamu Tsukamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier BiosciencesSuitaJapan
- The Osaka Medical Research Foundation for Intractable DiseasesOsakaJapan
- Department of Biochemistry, Hyogo College of MedicineNishinomiyaJapan
- Non-Profit Organization Think of Medicine in ScienceOsakaJapan
| | - Takuya Hasegawa
- Department of Cardiovascular Medicine, Garatia HospitalMinoJapan
| | - Ken Matsuoka
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier BiosciencesSuitaJapan
| | - Makoto Amaki
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular CenterSuitaJapan
| | - Hideaki Kanzaki
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular CenterSuitaJapan
| | - Chisato Izumi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular CenterSuitaJapan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier BiosciencesSuitaJapan
- The Osaka Medical Research Foundation for Intractable DiseasesOsakaJapan
- Non-Profit Organization Think of Medicine in ScienceOsakaJapan
| | - Shin Ito
- Department of Clinical Medicine and Development, National Cerebral and Cardiovascular CenterSuitaJapan
| | - Masafumi Kitakaze
- Department of Cardiovascular Medicine, Hanwa Memorial HospitalOsakaJapan
- The Osaka Medical Research Foundation for Intractable DiseasesOsakaJapan
- Non-Profit Organization Think of Medicine in ScienceOsakaJapan
| |
Collapse
|
17
|
Prickett TCR, Espiner EA, Pearson JF. Association of natriuretic peptides and receptor activity with cardio-metabolic health at middle age. Sci Rep 2024; 14:9919. [PMID: 38689031 PMCID: PMC11061163 DOI: 10.1038/s41598-024-60677-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024] Open
Abstract
Natriuretic peptides (NP) have multiple actions benefitting cardiovascular and metabolic health. Although many of these are mediated by Guanylyl Cyclase (GC) receptors NPR1 and NPR2, their role and relative importance in vivo is unclear. The intracellular mediator of NPR1 and NPR2, cGMP, circulates in plasma and can be used to examine relationships between receptor activity and tissue responses targeted by NPs. Plasma cGMP was measured in 348 participants previously recruited in a multidisciplinary community study (CHALICE) at age 50 years at a single centre. Associations between bio-active NPs and bio-inactive aminoterminal products with cGMP, and of cGMP with tissue response, were analysed using linear regression. Mediation of associations by NPs was assessed by Causal Mediation Analysis (CMA). ANP's contribution to cGMP far exceed those of other NPs. Modelling across three components (demographics, NPs and cardiovascular function) shows that ANP and CNP are independent and positive predictors of cGMP. Counter intuitively, findings from CMA imply that in specific tissues, NPR1 responds more to BNP stimulation than ANP. Collectively these findings align with longer tissue half-life of BNP, and direct further therapeutic interventions towards extending tissue activity of ANP and CNP.
Collapse
Affiliation(s)
- Timothy C R Prickett
- Departments of Medicine, University of Otago, Christchurch, PO Box 4345, Christchurch, 8140, New Zealand.
| | - Eric A Espiner
- Departments of Medicine, University of Otago, Christchurch, PO Box 4345, Christchurch, 8140, New Zealand
| | - John F Pearson
- Departments of Medicine, University of Otago, Christchurch, PO Box 4345, Christchurch, 8140, New Zealand
- Biostatistics and Computational Biology Unit, University of Otago, Christchurch, New Zealand
| |
Collapse
|
18
|
Cheslow L, Byrne M, Kopenhaver JS, Iacovitti L, Smeyne RJ, Snook AE, Waldman SA. GUCY2C signaling limits dopaminergic neuron vulnerability to toxic insults. NPJ Parkinsons Dis 2024; 10:83. [PMID: 38615030 PMCID: PMC11016112 DOI: 10.1038/s41531-024-00697-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 03/26/2024] [Indexed: 04/15/2024] Open
Abstract
Mitochondrial dysfunction and reactive oxygen species (ROS) accumulation within the substantia nigra pars compacta (SNpc) are central drivers of dopaminergic (DA) neuron death in Parkinson's disease (PD). Guanylyl cyclases and their second messenger cyclic (c)GMP support mitochondrial function, protecting against ROS and promoting cell survival in several tissues. However, the role of the guanylyl cyclase-cGMP axis in defining the vulnerability of DA neurons in the SNpc in PD remains unclear, in part due to the challenge of manipulating cGMP levels selectively in midbrain DA neurons. In that context, guanylyl cyclase C (GUCY2C), a receptor primarily expressed by intestinal epithelial cells, was discovered recently in midbrain DA neurons. Here, we demonstrate that GUCY2C promotes mitochondrial function, reducing oxidative stress and protecting DA neurons from degeneration in the 1-methyl-4-phenyl- 1,2,3,6-tetrahydropyridine (MPTP) mouse model. GUCY2C is overexpressed in the SNpc in PD patients and in mice treated with MPTP, possibly reflecting a protective response to oxidative stress. Moreover, cGMP signaling protects against oxidative stress, mitochondrial impairment, and cell death in cultured DA neurons. These observations reveal a previously unexpected role for the GUCY2C-cGMP signaling axis in controlling mitochondrial dysfunction and toxicity in SNpc DA neurons, highlighting the therapeutic potential of targeting DA neuron GUCY2C to prevent neurodegeneration in PD.
Collapse
Affiliation(s)
- Lara Cheslow
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Matthew Byrne
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jessica S Kopenhaver
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lorraine Iacovitti
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Richard J Smeyne
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam E Snook
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Microbiology & Immunology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Scott A Waldman
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Romero-Becera R, Santamans AM, Arcones AC, Sabio G. From Beats to Metabolism: the Heart at the Core of Interorgan Metabolic Cross Talk. Physiology (Bethesda) 2024; 39:98-125. [PMID: 38051123 DOI: 10.1152/physiol.00018.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/26/2023] [Accepted: 12/01/2023] [Indexed: 12/07/2023] Open
Abstract
The heart, once considered a mere blood pump, is now recognized as a multifunctional metabolic and endocrine organ. Its function is tightly regulated by various metabolic processes, at the same time it serves as an endocrine organ, secreting bioactive molecules that impact systemic metabolism. In recent years, research has shed light on the intricate interplay between the heart and other metabolic organs, such as adipose tissue, liver, and skeletal muscle. The metabolic flexibility of the heart and its ability to switch between different energy substrates play a crucial role in maintaining cardiac function and overall metabolic homeostasis. Gaining a comprehensive understanding of how metabolic disorders disrupt cardiac metabolism is crucial, as it plays a pivotal role in the development and progression of cardiac diseases. The emerging understanding of the heart as a metabolic and endocrine organ highlights its essential contribution to whole body metabolic regulation and offers new insights into the pathogenesis of metabolic diseases, such as obesity, diabetes, and cardiovascular disorders. In this review, we provide an in-depth exploration of the heart's metabolic and endocrine functions, emphasizing its role in systemic metabolism and the interplay between the heart and other metabolic organs. Furthermore, emerging evidence suggests a correlation between heart disease and other conditions such as aging and cancer, indicating that the metabolic dysfunction observed in these conditions may share common underlying mechanisms. By unraveling the complex mechanisms underlying cardiac metabolism, we aim to contribute to the development of novel therapeutic strategies for metabolic diseases and improve overall cardiovascular health.
Collapse
Affiliation(s)
| | | | - Alba C Arcones
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| |
Collapse
|
20
|
Ma X, McKie PM, Iyer SR, Scott C, Bailey K, Johnson BK, Benike SL, Chen H, Miller WL, Cabassi A, Burnett JC, Cannone V. MANP in Hypertension With Metabolic Syndrome: Proof-of-Concept Study of Natriuretic Peptide-Based Therapy for Cardiometabolic Disease. JACC Basic Transl Sci 2024; 9:18-29. [PMID: 38362338 PMCID: PMC10864980 DOI: 10.1016/j.jacbts.2023.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 02/17/2024]
Abstract
Hypertension and metabolic syndrome frequently coexist to increase the risk for adverse cardiometabolic outcomes. To date, no drug has been proven to be effective in treating hypertension with metabolic syndrome. M-atrial natriuretic peptide is a novel atrial natriuretic peptide analog that activates the particulate guanylyl cyclase A receptor. This study conducted a double-blind, placebo-controlled trial in 22 patients and demonstrated that a single subcutaneous injection of M-atrial natriuretic peptide was safe, well-tolerated, and exerted pleiotropic properties including blood pressure-lowering, lipolytic, and insulin resistance-improving effects. (MANP in Hypertension and Metabolic Syndrome [MANP-HTN-MS]; NCT03781739).
Collapse
Affiliation(s)
- Xiao Ma
- Cardiorenal Research Laboratory, Mayo Clinic, Rochester, Minnesota, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Paul M. McKie
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Seethalakshmi R. Iyer
- Cardiorenal Research Laboratory, Mayo Clinic, Rochester, Minnesota, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Christopher Scott
- Department of Health Science Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Kent Bailey
- Department of Health Science Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Bradley K. Johnson
- Department of Health Science Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Sherry L. Benike
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Horng Chen
- Cardiorenal Research Laboratory, Mayo Clinic, Rochester, Minnesota, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Wayne L. Miller
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Aderville Cabassi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - John C. Burnett
- Cardiorenal Research Laboratory, Mayo Clinic, Rochester, Minnesota, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Valentina Cannone
- Cardiorenal Research Laboratory, Mayo Clinic, Rochester, Minnesota, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
21
|
Shetty NS, Patel N, Gaonkar M, Li P, Arora G, Arora P. Natriuretic Peptide Normative Levels and Deficiency: The National Health and Nutrition Examination Survey. JACC. HEART FAILURE 2024; 12:50-63. [PMID: 37768244 PMCID: PMC10924765 DOI: 10.1016/j.jchf.2023.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Natriuretic peptides (NPs) are hormones with a range of key functions vital for cardiometabolic health. However, the reference ranges of NPs and the prevalence of NP deficiency in the healthy United States population remains poorly defined. OBJECTIVES This study aims to establish the reference range for N-terminal pro-B-type natriuretic peptide (NT-proBNP) values and to assess the prevalence of NP deficiency in a nationally representative healthy United States population. METHODS Healthy participants with NT-proBNP measurements from the 1999-2004 National Health and Nutrition Examination Survey were included. Weighted multivariable-adjusted linear regression models were used to assess the adjusted percentage difference of NT-proBNP concentrations by sex and race and ethnicity. NP deficiency was defined as concentrations <2.5th percentile in the study cohort. RESULTS Among 18,145 individuals (median age: 33.9 years [IQR: 17.1-49.0 years], 49.8% males, and 68.5% non-Hispanic White individuals), females had similar NT-proBNP concentrations in the 1-10 years group (4.2% [95% CI: -3.3% to 12.2%]), and highest differences in the 20-30 years group (150.5% [95% CI: 123.5%-180.8%]) compared with males in their respective age groups. Compared with non-Hispanic White individuals, non-Hispanic Black individuals had lower NT-proBNP concentrations in the 1- to 10-years group (19.6% [95% CI: 10.7%-27.6%]), and these differences were most pronounced in the 30-40 years group (40.2% [95% CI: 33.7%-46.0%]). An estimated 9.1 million United States individuals had NP deficiency. NP deficiency was associated with a higher risk of cardiometabolic diseases such as hypertension, dyslipidemia, obesity, and insulin resistance. CONCLUSIONS This study establishes the normative NP concentrations across the lifespan of a healthy United States population.
Collapse
Affiliation(s)
- Naman S Shetty
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nirav Patel
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mokshad Gaonkar
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Peng Li
- School of Nursing, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Garima Arora
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Pankaj Arora
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA; Section of Cardiology, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama, USA.
| |
Collapse
|
22
|
Ozkan B, Ndumele CE. Exploring the Mechanistic Link Between Obesity and Heart Failure. Curr Diab Rep 2023; 23:347-360. [PMID: 38100052 DOI: 10.1007/s11892-023-01526-y] [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] [Accepted: 11/07/2023] [Indexed: 01/14/2024]
Abstract
PURPOSE OF REVIEW Among subtypes of cardiovascular disease, obesity has a potent and unique association with heart failure (HF) that is unexplained by traditional cardiovascular risk mediators. The concomitant rise in the prevalence of obesity and HF necessitates better understanding of their relationship to develop effective prevention and treatment strategies. The purpose of this review is to provide mechanistic insight regarding the link between obesity and HF by elucidating the direct and indirect pathways linking the two conditions. RECENT FINDINGS Several direct pathophysiologic mechanisms contribute to HF risk in individuals with excess weight, including hemodynamic alterations, neurohormonal activation, hormonal effects of dysfunctional adipose tissue, ectopic fat deposition with resulting lipotoxicity and microvascular dysfunction. Obesity further predisposes to HF indirectly through causal associations with hypertension, dyslipidemia, and most importantly, diabetes via insulin resistance. Low levels of physical activity and fitness further influence HF risk in the context of obesity. These various processes lead to myocardial injury and cardiac remodeling that are reflected by abnormalities in cardiac biomarkers and cardiac function on myocardial imaging. Understanding and addressing obesity-associated HF is a pressing clinical and public health challenge which can be informed by a deeper understanding of the complex pathways linking these two conditions together.
Collapse
Affiliation(s)
- Bige Ozkan
- Division of Cardiology, Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, 600 N Wolfe St, Carnegie, Baltimore, MD, 568, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Chiadi E Ndumele
- Division of Cardiology, Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, 600 N Wolfe St, Carnegie, Baltimore, MD, 568, USA.
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
23
|
Jia B, Hasse A, Shi F, Collins S. Exercise performance is not improved in mice with skeletal muscle deletion of natriuretic peptide clearance receptor. PLoS One 2023; 18:e0293636. [PMID: 37917630 PMCID: PMC10621814 DOI: 10.1371/journal.pone.0293636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/16/2023] [Indexed: 11/04/2023] Open
Abstract
Natriuretic peptides (NP), including atrial, brain, and C-type natriuretic peptides (ANP, BNP, and CNP), play essential roles in regulating blood pressure, cardiovascular homeostasis, and systemic metabolism. One of the major metabolic effects of NP is manifested by their capacity to stimulate lipolysis and the thermogenesis gene program in adipocytes, however, in skeletal muscle their effects on metabolism and muscle function are not as well understood. There are three NP receptors (NPR): NPRA, NPRB, and NPRC, and all three NPR genes are expressed in skeletal muscle and C2C12 myocytes. In C2C12 myocytes treatment with either ANP, BNP, or CNP evokes the cGMP signaling pathway. Since NPRC functions as a clearance receptor and the amount of NPRC in a cell type determines the signaling strength of NPs, we generated a genetic model with Nprc gene deletion in skeletal muscle and tested whether enhancing NP signaling by preventing its clearance in skeletal muscle would improve exercise performance in mice. Under sedentary conditions, Nprc skeletal muscle knockout (MKO) mice showed comparable exercise performance to their floxed littermates in terms of maximal running velocity and total endurance running time. Eight weeks of voluntary running-wheel training in a young cohort significantly increased exercise performance, but no significant differences were observed in MKO compared with floxed control mice. Furthermore, 6-weeks of treadmill training in a relatively aged cohort also increased exercise performance compared with their baseline values, but again there were no differences between genotypes. In summary, our study suggests that NP signaling is potentially important in skeletal myocytes but its function in skeletal muscle in vivo needs to be further studied in additional physiological conditions or with new genetic mouse models.
Collapse
Affiliation(s)
- Brigitte Jia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Alexander Hasse
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Fubiao Shi
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL, United States of America
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL, United States of America
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville TN, United States of America
| |
Collapse
|
24
|
Espinoza-Derout J, Arambulo JML, Ramirez-Trillo W, Rivera JC, Hasan KM, Lao CJ, Jordan MC, Shao XM, Roos KP, Sinha-Hikim AP, Friedman TC. The lipolysis inhibitor acipimox reverses the cardiac phenotype induced by electronic cigarettes. Sci Rep 2023; 13:18239. [PMID: 37880325 PMCID: PMC10600141 DOI: 10.1038/s41598-023-44082-x] [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: 04/19/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023] Open
Abstract
Electronic cigarettes (e-cigarettes) are a prevalent alternative to conventional nicotine cigarettes among smokers and people who have never smoked. Increased concentrations of serum free fatty acids (FFAs) are crucial in generating lipotoxicity. We studied the effects of acipimox, an antilipolytic drug, on e-cigarette-induced cardiac dysfunction. C57BL/6J wild-type mice on high fat diet were treated with saline, e-cigarette with 2.4% nicotine [e-cigarette (2.4%)], and e-cigarette (2.4%) plus acipimox for 12 weeks. Fractional shortening and ejection fraction were diminished in mice exposed to e-cigarettes (2.4%) compared with saline and acipimox-treated mice. Mice exposed to e-cigarette (2.4%) had increased circulating levels of inflammatory cytokines and FFAs, which were diminished by acipimox. Gene Set Enrichment Analysis revealed that e-cigarette (2.4%)-treated mice had gene expression changes in the G2/M DNA damage checkpoint pathway that was normalized by acipimox. Accordingly, we showed that acipimox suppressed the nuclear localization of phospho-p53 induced by e-cigarette (2.4%). Additionally, e-cigarette (2.4%) increased the apurinic/apyrimidinic sites, a marker of oxidative DNA damage which was normalized by acipimox. Mice exposed to e-cigarette (2.4%) had increased cardiac Heme oxygenase 1 protein levels and 4-hydroxynonenal (4-HNE). These markers of oxidative stress were decreased by acipimox. Therefore, inhibiting lipolysis with acipimox normalizes the physiological changes induced by e-cigarettes and the associated increase in inflammatory cytokines, oxidative stress, and DNA damage.
Collapse
Affiliation(s)
- Jorge Espinoza-Derout
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA.
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Jose Mari Luis Arambulo
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
| | - William Ramirez-Trillo
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
| | - Juan Carlos Rivera
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
| | - Kamrul M Hasan
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Candice J Lao
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Maria C Jordan
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xuesi M Shao
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kenneth P Roos
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Amiya P Sinha-Hikim
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Theodore C Friedman
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
25
|
Cheslow L, Byrne M, Kopenhaver JS, Iacovitti L, Smeyne RJ, Snook AE, Waldman SA. GUCY2C signaling limits dopaminergic neuron vulnerability to toxic insults. RESEARCH SQUARE 2023:rs.3.rs-3416338. [PMID: 37886524 PMCID: PMC10602097 DOI: 10.21203/rs.3.rs-3416338/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Mitochondrial dysfunction and reactive oxygen species (ROS) accumulation within the substantia nigra pars compacta (SNpc) are central drivers of dopaminergic (DA) neuron death in Parkinson's disease (PD). Guanylyl cyclases, and their second messengers cyclic (c)GMP, support mitochondrial function, protecting against ROS and promoting cell survival in a number of tissues. However, the role of the guanylyl cyclase-cGMP axis in defining the vulnerability of DA neurons in the SNpc in PD remains unclear, in part due to the challenge of manipulating cGMP levels selectively in midbrain DA neurons. In that context, guanylyl cyclase C (GUCY2C), a receptor primarily expressed by intestinal epithelial cells, was discovered recently in midbrain DA neurons. Here, we demonstrate that GUCY2C promotes mitochondrial function, reducing oxidative stress and protecting DA neurons from degeneration in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of neurodegeneration. GUCY2C is overexpressed in the SNpc in PD patients and in mice treated with MPTP, possibly reflecting a protective response to oxidative stress. Moreover, cGMP signaling protects against oxidative stress, mitochondrial impairment, and cell death in cultured DA neurons. These observations reveal a previously unexpected role for the GUCY2C-cGMP signaling axis in controlling mitochondrial dysfunction and toxicity in nigral DA neurons, highlighting the therapeutic potential of targeting DA neuron GUCY2C to prevent neurodegeneration in PD.
Collapse
Affiliation(s)
- Lara Cheslow
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Matthew Byrne
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jessica S. Kopenhaver
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lorraine Iacovitti
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Richard J. Smeyne
- Department of Neurosciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam E. Snook
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Microbiology & Immunology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Scott A. Waldman
- Department of Pharmacology, Physiology, & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
26
|
Ichiki T, Cannone V, Scott CG, Iyer SR, Sangaralingham SJ, Bailey KR, Goetze JP, Tsuji Y, Rodeheffer RJ, Burnett JC. Sex-based differences in metabolic protection by the ANP genetic variant rs5068 in the general population. Am J Physiol Heart Circ Physiol 2023; 325:H545-H552. [PMID: 37417873 PMCID: PMC10538992 DOI: 10.1152/ajpheart.00321.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
Atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP) are produced in the heart and secreted into the circulation. As hormones, both peptides activate the guanylyl cyclase receptor A (GC-A), playing a role in blood pressure (BP) regulation. A significant role for ANP and BNP includes favorable actions in metabolic homeostasis. Sex-based high prevalence of risk factors for cardiovascular disease in males compared with females is well established, but sex-based differences on cardiometabolic protection have not been investigated in relation to ANP (NPPA) and BNP (NPPB) gene variants. We included 1,146 subjects in the general population from Olmsted County, Minnesota. Subjects were genotyped for the ANP gene variant rs5068 and BNP gene variant rs198389. Cardiometabolic parameters and medical records were reviewed. In the presence of the minor allele of rs5068, diastolic BP, creatinine, body mass index (BMI), waist measurement, insulin, and prevalence of obesity and metabolic syndrome were lower, whereas HDL was higher in males with only trends observed in females. We observed no associations of the minor allele with echocardiographic parameters in either males or females. Regarding rs198389 genotype, the minor allele was not associated with any BP, metabolic, renal, or echocardiographic parameters in either sex. In the general community, the minor allele of the ANP gene variant rs5068 is associated with a favorable metabolic phenotype in males. No associations were observed with the BNP gene variant rs198389. These studies support a protective role of the ANP pathway on metabolic function and underscore the importance of sex in relationship to natriuretic peptide responses.NEW & NOTEWORTHY Males are characterized by lower ANP and BNP with greater prevalence of cardiometabolic disease. The ANP genetic variant rs5068 was associated with less metabolic dysfunction in males, whereas no metabolic profile was related to the BNP genetic variant rs198389 in the general population. ANP may play a more biological role in metabolic homeostasis compared with BNP in the general population with greater physiological metabolic actions in males compared with females.
Collapse
Affiliation(s)
- Tomoko Ichiki
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Community and General Medicine, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Valentina Cannone
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Christopher G Scott
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States
| | - Seethalakshmi R Iyer
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Kent R Bailey
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States
| | - Jens P Goetze
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Yoshihisa Tsuji
- Department of Community and General Medicine, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Richard J Rodeheffer
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
27
|
Echouffo-Tcheugui JB, Zhang S, McEvoy JW, Juraschek SP, Coresh J, Christenson RH, Ndumele CE, Selvin E. Body Composition Measures and N-terminal pro-B-type Natriuretic Peptide (NT-pro-BNP) in US Adults. Clin Chem 2023; 69:901-914. [PMID: 37477552 PMCID: PMC10478300 DOI: 10.1093/clinchem/hvad085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/10/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND The associations of N-terminal pro-B-type natriuretic peptide (NT-pro-BNP) with dual energy x-ray absorptiometry (DEXA)-derived measures of body mass and composition are largely unknown. METHODS We included participants aged ≥20 years from the 1999-2004 National Health and Nutrition Examination Survey with NT-pro-BNP and DEXA-derived body composition (fat and lean mass) measures. We used linear and logistic regression to characterize the associations of measures of body mass and composition (body mass index [BMI], waist circumference [WC], fat mass, and lean mass) with NT-pro-BNP, adjusting for cardiovascular risk factors. RESULTS We conducted sex-specific analyses among 9134 adults without cardiovascular disease (mean age 44.4 years, 50.8% women, and 72% White adults). The adjusted mean NT-proBNP values were lowest in the highest quartiles of BMI, WC, fat mass, and lean mass. There were large adjusted absolute differences in NT-pro-BNP between the highest and lowest quartiles of DEXA-derived lean mass, -6.26 pg/mL (95% confidence interval [CI], -8.99 to -3.52) among men and -22.96 pg/mL (95% CI, -26.83 to -19.09) among women. Lean mass exhibited a strong inverse association with elevated NT-pro-BNP ≥ 81.4 pg/mL (highest quartile) - odds ratio (OR) 0.58 (95% CI, 0.39-0.86) in men and OR 0.59 (95% CI, 0.47-0.73) in women for highest lean mass quartile vs. lowest quartile. Further adjustment for fat mass, BMI, or WC did not appreciably alter the inverse association of lean mass with NT-pro-BNP. CONCLUSIONS In a national sample of US adults, lean mass was inversely associated with NT-pro-BNP.
Collapse
Affiliation(s)
- Justin B. Echouffo-Tcheugui
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Sui Zhang
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - John W. McEvoy
- Division of Cardiology and National Institute for Prevention and Cardiovascular Health, National University of Ireland, Galway, Ireland
| | - Stephen P. Juraschek
- Division of General Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Josef Coresh
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Robert H. Christenson
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Chiadi E. Ndumele
- Division of Cardiology, Department of Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Elizabeth Selvin
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
28
|
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
|
29
|
Świeżawska-Boniecka B, Szmidt-Jaworska A. Phytohormones and cyclic nucleotides - Long-awaited couples? JOURNAL OF PLANT PHYSIOLOGY 2023; 286:154005. [PMID: 37186984 DOI: 10.1016/j.jplph.2023.154005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/09/2023] [Indexed: 05/17/2023]
Affiliation(s)
- Brygida Świeżawska-Boniecka
- Nicolaus Copernicus University, Faculty of Biological and Veterinary Sciences, Department of Plant Physiology and Biotechnology, Lwowska St. 1, PL 87-100, Torun, Poland.
| | - Adriana Szmidt-Jaworska
- Nicolaus Copernicus University, Faculty of Biological and Veterinary Sciences, Department of Plant Physiology and Biotechnology, Lwowska St. 1, PL 87-100, Torun, Poland.
| |
Collapse
|
30
|
Wang L, Tang Y, Herman MA, Spurney RF. Pharmacologic blockade of the natriuretic peptide clearance receptor promotes weight loss and enhances insulin sensitivity in type 2 diabetes. Transl Res 2023; 255:140-151. [PMID: 36563959 PMCID: PMC10441142 DOI: 10.1016/j.trsl.2022.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/30/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
While natriuretic peptides (NPs) are primarily known for their renal and cardiovascular actions, NPs stimulate lipolysis in adipocytes and induce a thermogenic program in white adipose tissue (WAT) that resembles brown fat. The biologic effects of NPs are negatively regulated by the NP clearance receptor (NPRC), which binds and degrades NPs. Knockout (KO) of NPRC protects against diet induced obesity and improves insulin sensitivity in obese mice. To determine if pharmacologic blockade of NPRC enhanced the beneficial metabolic actions of NPs in type 2 diabetes, we blocked NP clearance in a mouse model of type 2 diabetes using the specific NPRC ligand ANP(4-23). We found that treatment with ANP(4-23) caused a significant decrease in body weight by increasing energy expenditure and reducing fat mass without a change in lean body mass. The decrease in fat mass was associated with a significant improvement in insulin sensitivity and reduced serum insulin levels. These beneficial effects were accompanied by a decrease in infiltrating macrophages in adipose tissue, and reduced expression of inflammatory markers in both serum and WAT. These data suggest that inhibiting NP clearance may be an effective pharmacologic approach to promote weight loss and enhance insulin sensitivity in type 2 diabetes. Optimizing the therapeutic approach may lead to useful therapies for obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Liming Wang
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| | - Yuping Tang
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| | - Mark A Herman
- Division of Endocrinology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina; Duke Molecular Physiology Institute, Durham, North Carolina
| | - Robert F Spurney
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina.
| |
Collapse
|
31
|
Chang P, Zhang X, Zhang J, Wang J, Wang X, Li M, Wang R, Yu J, Fu F. BNP protects against diabetic cardiomyopathy by promoting Opa1-mediated mitochondrial fusion via activating the PKG-STAT3 pathway. Redox Biol 2023; 62:102702. [PMID: 37116257 PMCID: PMC10165144 DOI: 10.1016/j.redox.2023.102702] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/25/2023] [Accepted: 04/14/2023] [Indexed: 04/30/2023] Open
Abstract
Brain natriuretic peptide (BNP) belongs to the family of natriuretic peptides, which are responsible for a wide range of actions. Diabetic cardiomyopathy (DCM) is often associated with increased BNP levels. This present research intends to explore the role of BNP in the development of DCM and the underlying mechanisms. Diabetes was induced in mice using streptozotocin (STZ). Primary neonatal cardiomyocytes were treated with high glucose. It was found that the levels of plasma BNP started to increase at 8 weeks after diabetes, which preceded the development of DCM. Addition of exogenous BNP promoted Opa1-mediated mitochondrial fusion, inhibited mitochondrial oxidative stress, preserved mitochondrial respiratory capacity and prevented the development of DCM, while knockdown of endogenous BNP exacerbated mitochondrial dysfunction and accelerated DCM. Opa1 knockdown attenuated the aforementioned protective action of BNP both in vivo and in vitro. BNP-induced mitochondrial fusion requires the activation of STAT3, which facilitated Opa1 transcription by binding to its promoter regions. PKG, a crucial signaling biomolecule in the BNP signaling pathway, interacted with STAT3 and induced its activation. Knockdown of NPRA (the receptor of BNP) or PKG blunted the promoting effect of BNP on STAT3 phosphorylation and Opa1-mediated mitochondrial fusion. The results of this study demonstrate for the first time that there is a rise in BNP during the early stages of DCM as a compensatory protection mechanism. BNP is a novel mitochondrial fusion activator in protecting against hyperglycemia-induced mitochondrial oxidative injury and DCM through the activation of NPRA-PKG-STAT3-Opa1 signaling pathway.
Collapse
Affiliation(s)
- Pan Chang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, 710038, China; Clinical Experimental Center, The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710100, China
| | - Xiaomeng Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Jing Zhang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, 710038, China
| | - Jianbang Wang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, 710038, China
| | - Xihui Wang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, 710038, China
| | - Man Li
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, 710038, China; Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, Xi'an, 710032, China
| | - Rui Wang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, 710038, China
| | - Jun Yu
- Clinical Experimental Center, The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710100, China.
| | - Feng Fu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, Xi'an, 710032, China; Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710038, China.
| |
Collapse
|
32
|
Harris MP, Zeng S, Zhu Z, Lira VA, Yu L, Hodgson-Zingman DM, Zingman LV. Myokine Musclin Is Critical for Exercise-Induced Cardiac Conditioning. Int J Mol Sci 2023; 24:6525. [PMID: 37047496 PMCID: PMC10095193 DOI: 10.3390/ijms24076525] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
This study investigates the role and mechanisms by which the myokine musclin promotes exercise-induced cardiac conditioning. Exercise is one of the most powerful triggers of cardiac conditioning with proven benefits for healthy and diseased hearts. There is an emerging understanding that muscles produce and secrete myokines, which mediate local and systemic "crosstalk" to promote exercise tolerance and overall health, including cardiac conditioning. The myokine musclin, highly conserved across animal species, has been shown to be upregulated in response to physical activity. However, musclin effects on exercise-induced cardiac conditioning are not established. Following completion of a treadmill exercise protocol, wild type (WT) mice and mice with disruption of the musclin-encoding gene, Ostn, had their hearts extracted and exposed to an ex vivo ischemia-reperfusion protocol or biochemical studies. Disruption of musclin signaling abolished the ability of exercise to mitigate cardiac ischemic injury. This impaired cardioprotection was associated with reduced mitochondrial content and function linked to blunted cyclic guanosine monophosphate (cGMP) signaling. Genetic deletion of musclin reduced the nuclear abundance of protein kinase G (PKGI) and cyclic adenosine monophosphate (cAMP) response element binding (CREB), resulting in suppression of the master regulator of mitochondrial biogenesis, peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α), and its downstream targets in response to physical activity. Synthetic musclin peptide pharmacokinetic parameters were defined and used to calculate the infusion rate necessary to maintain its plasma level comparable to that observed after exercise. This infusion was found to reproduce the cardioprotective benefits of exercise in sedentary WT and Ostn-KO mice. Musclin is essential for exercise-induced cardiac protection. Boosting musclin signaling might serve as a novel therapeutic strategy for cardioprotection.
Collapse
Affiliation(s)
- Matthew P. Harris
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Center, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Shemin Zeng
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Center, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA
- Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| | - Zhiyong Zhu
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Center, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA
- Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| | - Vitor A. Lira
- Department of Health and Human Physiology, Fraternal Order of Eagles Diabetes Center, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Liping Yu
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Center, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA
- NMR Core Facility and Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA
| | - Denice M. Hodgson-Zingman
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Center, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Leonid V. Zingman
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Center, Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA
- Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| |
Collapse
|
33
|
Anitha A, Thanseem I, Iype M, Thomas SV. Mitochondrial dysfunction in cognitive neurodevelopmental disorders: Cause or effect? Mitochondrion 2023; 69:18-32. [PMID: 36621534 DOI: 10.1016/j.mito.2023.01.002] [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: 09/24/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
Mitochondria have a crucial role in brain development and neurogenesis, both in embryonic and adult brains. Since the brain is the highest energy consuming organ, it is highly vulnerable to mitochondrial dysfunction. This has been implicated in a range of brain disorders including, neurodevelopmental conditions, psychiatric illnesses, and neurodegenerative diseases. Genetic variations in mitochondrial DNA (mtDNA), and nuclear DNA encoding mitochondrial proteins, have been associated with several cognitive disorders. However, it is not yet clear whether mitochondrial dysfunction is a primary cause of these conditions or a secondary effect. Our review article deals with this topic, and brings out recent advances in mitochondria-oriented therapies. Mitochondrial dysfunction could be involved in the pathogenesis of a subset of disorders involving cognitive impairment. In these patients, mitochondrial dysfunction could be the cause of the condition, rather than the consequence. There are vast areas in this topic that remains to be explored and elucidated.
Collapse
Affiliation(s)
- Ayyappan Anitha
- Dept. of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Shoranur, Palakkad 679 523, Kerala, India.
| | - Ismail Thanseem
- Dept. of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Shoranur, Palakkad 679 523, Kerala, India
| | - Mary Iype
- Dept. of Pediatric Neurology, Government Medical College, Thiruvananthapuram 695 011, Kerala, India; Dept. of Neurology, ICCONS, Thiruvananthapuram 695 033, Kerala, India
| | - Sanjeev V Thomas
- Dept. of Neurology, ICCONS, Thiruvananthapuram 695 033, Kerala, India
| |
Collapse
|
34
|
Volpe M, Gallo G, Rubattu S. Endocrine functions of the heart: from bench to bedside. Eur Heart J 2023; 44:643-655. [PMID: 36582126 DOI: 10.1093/eurheartj/ehac759] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/22/2022] [Accepted: 12/05/2022] [Indexed: 12/31/2022] Open
Abstract
Heart has a recognized endocrine function as it produces several biologically active substances with hormonal properties. Among these hormones, the natriuretic peptide (NP) system has been extensively characterized and represents a prominent expression of the endocrine function of the heart. Over the years, knowledge about the mechanisms governing their synthesis, secretion, processing, and receptors interaction of NPs has been intensively investigated. Their main physiological endocrine and paracrine effects on cardiovascular and renal systems are mostly mediated through guanylate cyclase-A coupled receptors. The potential role of NPs in the pathophysiology of heart failure and particularly their counterbalancing action opposing the overactivation of renin-angiotensin-aldosterone and sympathetic nervous systems has been described. In addition, NPs are used today as key biomarkers in cardiovascular diseases with both diagnostic and prognostic significance. On these premises, multiple therapeutic strategies based on the biological properties of NPs have been attempted to develop new cardiovascular therapies. Apart from the introduction of the class of angiotensin receptor/neprilysin inhibitors in the current management of heart failure, novel promising molecules, including M-atrial natriuretic peptide (a novel atrial NP-based compound), have been tested for the treatment of human hypertension. The development of new drugs is currently underway, and we are probably only at the dawn of novel NPs-based therapeutic strategies. The present article also provides an updated overview of the regulation of NPs synthesis and secretion by microRNAs and epigenetics as well as interactions of cardiac hormones with other endocrine systems.
Collapse
Affiliation(s)
- Massimo Volpe
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy.,IRCCS San Raffaele, Via della Pisana 235, 00163 Rome, Italy
| | - Giovanna Gallo
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy
| | - Speranza Rubattu
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy.,IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli (IS), Italy
| |
Collapse
|
35
|
Shan S, Qiao Q, Yin R, Zhang L, Shi J, Zhao W, Zhou J, Li Z. Identification of a Novel Strain Lactobacillus Reuteri and Anti-Obesity Effect through Metabolite Indole-3-Carboxaldehyde in Diet-Induced Obese Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:3239-3249. [PMID: 36786753 DOI: 10.1021/acs.jafc.2c05764] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The potentially beneficial effects of probiotics in the treatment of obesity have been generally demonstrated. In the present study, a new strain of Lactobacillus reuteri SY523 (L. reuteri SY523) with an anti-obesity effect was isolated from the fecal microbiota of diet-induced obese mice. Untargeted metabolomics analysis of mice serum showed that the significantly differential metabolite indole-3-carboxaldehyde (3-IAId) was markedly elevated in the L. reuteri SY523-treated group, and interestingly, the abundance of 3-IAId was significantly negatively associated with obesity-related indicators. As expected, in the HepG2 cell induced by free fatty acids, the potential activity of 3-IAId in restraining lipid deposition was verified. Further, we found that 3-IAId was involved in the anti-obesity effect of L. reuteri SY523 mainly via regulating the cGMP/cAMP signaling pathway. The highlight of this study lies in clarifying the pivotal role of metabolite 3-IAId in the anti-obesity effect induced by L. reuteri SY523, which is conducive to the development of probiotics for anti-obesity agents.
Collapse
Affiliation(s)
- Shuhua Shan
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Qinqin Qiao
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Ruopeng Yin
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lizhen Zhang
- School of Life Science, Shanxi University, Taiyuan 030006, China
| | - Jiangying Shi
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Wenjing Zhao
- Department of Biology, Taiyuan Normal University, Jinzhong 030619, China
| | - Jiaqi Zhou
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Zhuoyu Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
36
|
Gerovska D, Araúzo-Bravo MJ. Skeletal Muscles of Sedentary and Physically Active Aged People Have Distinctive Genic Extrachromosomal Circular DNA Profiles. Int J Mol Sci 2023; 24:ijms24032736. [PMID: 36769072 PMCID: PMC9917053 DOI: 10.3390/ijms24032736] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
To bring new extrachromosomal circular DNA (eccDNA) enrichment technologies closer to the clinic, specifically for screening, early diagnosis, and monitoring of diseases or lifestyle conditions, it is paramount to identify the differential pattern of the genic eccDNA signal between two states. Current studies using short-read sequenced purified eccDNA data are based on absolute numbers of unique eccDNAs per sample or per gene, length distributions, or standard methods for RNA-seq differential analysis. Previous analyses of RNA-seq data found significant transcriptomics difference between sedentary and active life style skeletal muscle (SkM) in young people but very few in old. The first attempt using circulomics data from SkM and blood of aged lifelong sedentary and physically active males found no difference at eccDNA level. To improve the capability of finding differences between circulomics data groups, we designed a computational method to identify Differentially Produced per Gene Circles (DPpGCs) from short-read sequenced purified eccDNA data based on the circular junction, split-read signal, of the eccDNA, and implemented it into a software tool DifCir in Matlab. We employed DifCir to find to the distinctive features of the influence of the physical activity or inactivity in the aged SkM that would have remained undetected by transcriptomics methods. We mapped the data from tissue from SkM and blood from two groups of aged lifelong sedentary and physically active males using Circle_finder and subsequent merging and filtering, to find the number and length distribution of the unique eccDNA. Next, we used DifCir to find up-DPpGCs in the SkM of the sedentary and active groups. We assessed the functional enrichment of the DPpGCs using Disease Gene Network and Gene Set Enrichment Analysis. To find genes that produce eccDNA in a group without comparison with another group, we introduced a method to find Common PpGCs (CPpGCs) and used it to find CPpGCs in the SkM of the sedentary and active group. Finally, we found the eccDNA that carries whole genes. We discovered that the eccDNA in the SkM of the sedentary group is not statistically different from that of physically active aged men in terms of number and length distribution of eccDNA. In contrast, with DifCir we found distinctive gene-associated eccDNA fingerprints. We identified statistically significant up-DPpGCs in the two groups, with the top up-DPpGCs shed by the genes AGBL4, RNF213, DNAH7, MED13, and WWTR1 in the sedentary group, and ZBTB7C, TBCD, ITPR2, and DDX11-AS1 in the active group. The up-DPpGCs in both groups carry mostly gene fragments rather than whole genes. Though the subtle transcriptomics difference, we found RYR1 to be both transcriptionally up-regulated and up-DPpGCs gene in sedentary SkM. DifCir emphasizes the high sensitivity of the circulome compared to the transcriptome to detect the molecular fingerprints of exercise in aged SkM. It allows efficient identification of gene hotspots that excise more eccDNA in a health state or disease compared to a control condition.
Collapse
Affiliation(s)
- Daniela Gerovska
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, Calle Doctor Begiristain s/n, 20014 San Sebastian, Spain
- Correspondence: (D.G.); (M.J.A.-B.)
| | - Marcos J. Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, Calle Doctor Begiristain s/n, 20014 San Sebastian, Spain
- Basque Foundation for Science, IKERBASQUE, Calle María Díaz Harokoa 3, 48013 Bilbao, Spain
- CIBER of Frailty and Healthy Aging (CIBERfes), 28029 Madrid, Spain
- Max Planck Institute for Molecular Biomedicine, Computational Biology and Bioinformatics, Röntgenstr. 20, 48149 Münster, Germany
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), 48940 Leioa, Spain
- Correspondence: (D.G.); (M.J.A.-B.)
| |
Collapse
|
37
|
Horikoshi T, Nakamura T, Yamaguchi K, Yoshizaki T, Watanabe Y, Kuroki K, Uematsu M, Nakamura K, Kobayashi T, Sato A. Prognostic Value of Novel Natriuretic Peptide Index After Percutaneous Coronary Intervention. Circ J 2023; 87:296-305. [PMID: 36261336 DOI: 10.1253/circj.cj-22-0531] [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] [Indexed: 01/31/2023]
Abstract
BACKGROUND The predictive value of both atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP) is well known. This study evaluated the prognostic value of a novel natriuretic peptide index (NPI) combining ANP and BNP. METHODS AND RESULTS This study included 849 consecutive patients with coronary artery disease who underwent successful percutaneous coronary intervention (PCI). Patients were followed up clinically for up to 3 years or until the occurrence of major adverse cardiac events (MACE). The primary endpoint was a composite of all-cause death and non-fatal myocardial infarction. The NPI (pg/mL) was defined as √ANP×BNP. MACE occurred in 73 patients (8.6%) during the follow-up period. Receiver operating characteristic curve analysis showed the highest area under the curve for NPI (0.779) compared with ANP and BNP (0.773 and 0.755, respectively). A risk analysis of MACE occurrence adjusted for the multivariable model showed the highest hazard ratio (HR) for NPI (1.33; 95% confidence interval [CI] 1.18-1.51; P<0.001) compared with ANP and BNP (HR 1.25 [95% CI 1.13-1.39] and 1.30 [95% CI 1.13-1.49], respectively; P<0.001). The NPI was a significant independent predictor of MACE, among other clinical parameters, in the multivariable analysis. CONCLUSIONS Compared with ANP and BNP, the NPI was more effective in predicting future adverse events after PCI.
Collapse
Affiliation(s)
- Takeo Horikoshi
- Department of Cardiology, University of Yamanashi, Faculty of Medicine
| | | | | | - Toru Yoshizaki
- Department of Cardiology, University of Yamanashi, Faculty of Medicine
| | - Yosuke Watanabe
- Department of Cardiology, University of Yamanashi, Faculty of Medicine
| | - Kenji Kuroki
- Department of Cardiology, University of Yamanashi, Faculty of Medicine
| | - Manabu Uematsu
- Department of Cardiology, University of Yamanashi, Faculty of Medicine
| | - Kazuto Nakamura
- Department of Cardiology, University of Yamanashi, Faculty of Medicine
| | | | - Akira Sato
- Department of Cardiology, University of Yamanashi, Faculty of Medicine
| |
Collapse
|
38
|
Samidurai A, Xi L, Das A, Kukreja RC. Beyond Erectile Dysfunction: cGMP-Specific Phosphodiesterase 5 Inhibitors for Other Clinical Disorders. Annu Rev Pharmacol Toxicol 2023; 63:585-615. [PMID: 36206989 DOI: 10.1146/annurev-pharmtox-040122-034745] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cyclic guanosine monophosphate (cGMP), an important intracellular second messenger, mediates cellular functional responses in all vital organs. Phosphodiesterase 5 (PDE5) is one of the 11 members of the cyclic nucleotide phosphodiesterase (PDE) family that specifically targets cGMP generated by nitric oxide-driven activation of the soluble guanylyl cyclase. PDE5 inhibitors, including sildenafil and tadalafil, are widely used for the treatment of erectile dysfunction, pulmonary arterial hypertension, and certain urological disorders. Preclinical studies have shown promising effects of PDE5 inhibitors in the treatment of myocardial infarction, cardiac hypertrophy, heart failure, cancer and anticancer-drug-associated cardiotoxicity, diabetes, Duchenne muscular dystrophy, Alzheimer's disease, and other aging-related conditions. Many clinical trials with PDE5 inhibitors have focused on the potential cardiovascular, anticancer, and neurological benefits. In this review, we provide an overview of the current state of knowledge on PDE5 inhibitors and their potential therapeutic indications for various clinical disorders beyond erectile dysfunction.
Collapse
Affiliation(s)
- Arun Samidurai
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| | - Lei Xi
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| | - Rakesh C Kukreja
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| |
Collapse
|
39
|
Werbner B, Tavakoli-Rouzbehani OM, Fatahian AN, Boudina S. The dynamic interplay between cardiac mitochondrial health and myocardial structural remodeling in metabolic heart disease, aging, and heart failure. THE JOURNAL OF CARDIOVASCULAR AGING 2023; 3:9. [PMID: 36742465 PMCID: PMC9894375 DOI: 10.20517/jca.2022.42] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This review provides a holistic perspective on the bi-directional relationship between cardiac mitochondrial dysfunction and myocardial structural remodeling in the context of metabolic heart disease, natural cardiac aging, and heart failure. First, a review of the physiologic and molecular drivers of cardiac mitochondrial dysfunction across a range of increasingly prevalent conditions such as metabolic syndrome and cardiac aging is presented, followed by a general review of the mechanisms of mitochondrial quality control (QC) in the heart. Several important mechanisms by which cardiac mitochondrial dysfunction triggers or contributes to structural remodeling of the heart are discussed: accumulated metabolic byproducts, oxidative damage, impaired mitochondrial QC, and mitochondrial-mediated cell death identified as substantial mechanistic contributors to cardiac structural remodeling such as hypertrophy and myocardial fibrosis. Subsequently, the less studied but nevertheless important reverse relationship is explored: the mechanisms by which cardiac structural remodeling feeds back to further alter mitochondrial bioenergetic function. We then provide a condensed pathogenesis of several increasingly important clinical conditions in which these relationships are central: diabetic cardiomyopathy, age-associated declines in cardiac function, and the progression to heart failure, with or without preserved ejection fraction. Finally, we identify promising therapeutic opportunities targeting mitochondrial function in these conditions.
Collapse
Affiliation(s)
- Benjamin Werbner
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Amir Nima Fatahian
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Sihem Boudina
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
40
|
Welsh P, Campbell RT, Mooney L, Kimenai DM, Hayward C, Campbell A, Porteous D, Mills NL, Lang NN, Petrie MC, Januzzi JL, McMurray JJ, Sattar N. Reference Ranges for NT-proBNP (N-Terminal Pro-B-Type Natriuretic Peptide) and Risk Factors for Higher NT-proBNP Concentrations in a Large General Population Cohort. Circ Heart Fail 2022; 15:e009427. [PMID: 36098049 PMCID: PMC9561238 DOI: 10.1161/circheartfailure.121.009427] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 07/11/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Demographic differences in expected NT-proBNP (N-terminal pro-B-type natriuretic peptide) concentration are not well established. We aimed to establish reference ranges for NT-proBNP and explore the determinants of moderately elevated NT-proBNP under the universal definition of heart failure criteria. METHODS This is a cross-sectional study. NT-proBNP was measured in serum from 18 356 individuals without previous cardiovascular disease in the Generation Scotland Scottish Family Health Study. Age- and sex-stratified medians and 97.5th centiles were generated. Sex stratified risk factors for moderately elevated NT-proBNP (≥125 pg/mL) were investigated. RESULTS In males, median (97.5th centile) NT-proBNP concentration at age <30 years was 21 (104) pg/mL, rising to 38 (195) pg/ml at 50 to 59 years, and 281 (6792) pg/mL at ≥80 years. In females, median NT-proBNP at age <30 years was 51 (196) pg/mL, 66 (299) pg/mL at 50 to 59 years, and 240 (2704) pg/mL at ≥80 years. At age <30 years, 9.8% of females and 1.4% of males had elevated NT-proBNP, rising to 76.5% and 81.0%, respectively, at age ≥80 years. After adjusting for risk factors, an NT-proBNP ≥125 pg/mL was more common in females than males (OR, 9.48 [95% CI, 5.60-16.1]). Older age and smoking were more strongly associated with elevated NT-proBNP in males than in females (Psex interaction <0.001, 0.07, respectively). Diabetes was inversely associated with odds of elevated NT-proBNP in females only (Psex interaction=0.007). CONCLUSIONS An NT-proBNP ≥125 pg/mL is common in females without classical cardiovascular risk factors as well as older people. If NT-proBNP becomes widely used for screening in the general population, interpretation of NT-proBNP levels will require that age and sex-specific thresholds are used to identify patients with potential pathophysiology.
Collapse
Affiliation(s)
- Paul Welsh
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom (P.W., R.T.C., L.M., N.N.L., M.C.P., J.J.V.M., N.S.)
| | - Ross T. Campbell
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom (P.W., R.T.C., L.M., N.N.L., M.C.P., J.J.V.M., N.S.)
| | - Leanne Mooney
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom (P.W., R.T.C., L.M., N.N.L., M.C.P., J.J.V.M., N.S.)
| | - Dorien M. Kimenai
- British Heart Foundation Centre for Cardiovascular Science (D.M.K., N.L.M.), University of Edinburgh, United Kingdom
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit (C.H.), University of Edinburgh, United Kingdom
| | - Archie Campbell
- Institute of Genetics and Cancer (A.C., D.P.), University of Edinburgh, United Kingdom
| | - David Porteous
- Institute of Genetics and Cancer (A.C., D.P.), University of Edinburgh, United Kingdom
| | - Nicholas L. Mills
- British Heart Foundation Centre for Cardiovascular Science (D.M.K., N.L.M.), University of Edinburgh, United Kingdom
- Usher Institute (N.L.M.), University of Edinburgh, United Kingdom
| | - Ninian N. Lang
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom (P.W., R.T.C., L.M., N.N.L., M.C.P., J.J.V.M., N.S.)
| | - Mark C. Petrie
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom (P.W., R.T.C., L.M., N.N.L., M.C.P., J.J.V.M., N.S.)
| | - James L. Januzzi
- Cardiology Division, Massachusetts General Hospital, Boston (J.L.J.)
- Harvard Medical School, Boston, MA (J.L.J.)
| | - John J.V. McMurray
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom (P.W., R.T.C., L.M., N.N.L., M.C.P., J.J.V.M., N.S.)
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom (P.W., R.T.C., L.M., N.N.L., M.C.P., J.J.V.M., N.S.)
| |
Collapse
|
41
|
Exogenous ANP Treatment Ameliorates Myocardial Insulin Resistance and Protects against Ischemia-Reperfusion Injury in Diet-Induced Obesity. Int J Mol Sci 2022; 23:ijms23158373. [PMID: 35955507 PMCID: PMC9369294 DOI: 10.3390/ijms23158373] [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: 07/04/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023] Open
Abstract
Increasing evidence suggests natriuretic peptides (NPs) coordinate interorgan metabolic crosstalk. We recently reported exogenous ANP treatment ameliorated systemic insulin resistance by inducing adipose tissue browning and attenuating hepatic steatosis in diet-induced obesity (DIO). We herein investigated whether ANP treatment also ameliorates myocardial insulin resistance, leading to cardioprotection during ischemia-reperfusion injury (IRI) in DIO. Mice fed a high-fat diet (HFD) or normal-fat diet for 13 weeks were treated with or without ANP infusion subcutaneously for another 3 weeks. Left ventricular BNP expression was substantially reduced in HFD hearts. Intraperitoneal-insulin-administration-induced Akt phosphorylation was impaired in HFD hearts, which was restored by ANP treatment, suggesting that ANP treatment ameliorated myocardial insulin resistance. After ischemia-reperfusion using the Langendorff model, HFD impaired cardiac functional recovery with a corresponding increased infarct size. However, ANP treatment improved functional recovery and reduced injury while restoring impaired IRI-induced Akt phosphorylation in HFD hearts. Myocardial ultrastructural analyses showed increased peri-mitochondrial lipid droplets with concomitantly decreased ATGL and HSL phosphorylation levels in ANP-treated HFD, suggesting that ANP protects mitochondria from lipid overload by trapping lipids. Accordingly, ANP treatment attenuated mitochondria cristae disruption after IRI in HFD hearts. In summary, exogenous ANP treatment ameliorates myocardial insulin resistance and protects against IRI associated with mitochondrial ultrastructure modifications in DIO. Replenishing biologically active NPs substantially affects HFD hearts in which endogenous NP production is impaired.
Collapse
|
42
|
Zhang X, Li W, Zhou T, Liu M, Wu Q, Dong N. Corin Deficiency Alters Adipose Tissue Phenotype and Impairs Thermogenesis in Mice. BIOLOGY 2022; 11:biology11081101. [PMID: 35892957 PMCID: PMC9329919 DOI: 10.3390/biology11081101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022]
Abstract
Atrial natriuretic peptide (ANP) is a key regulator in body fluid balance and cardiovascular biology. In addition to its role in enhancing natriuresis and vasodilation, ANP increases lipolysis and thermogenesis in adipose tissue. Corin is a protease responsible for ANP activation. It remains unknown if corin has a role in regulating adipose tissue function. Here, we examined adipose tissue morphology and function in corin knockout (KO) mice. We observed increased weights and cell sizes in white adipose tissue (WAT), decreased levels of uncoupling protein 1 (Ucp1), a brown adipocyte marker in WAT and brown adipose tissue (BAT), and suppressed thermogenic gene expression in BAT from corin KO mice. At regular room temperature, corin KO and wild-type mice had similar metabolic rates. Upon cold exposure at 4 °C, corin KO mice exhibited impaired thermogenic responses and developed hypothermia. In BAT from corin KO mice, the signaling pathway of p38 mitogen-activated protein kinase, peroxisome proliferator-activated receptor c coactivator 1a, and Ucp1 was impaired. In cell culture, ANP treatment increased Ucp1 expression in BAT-derived adipocytes from corin KO mice. These data indicate that corin mediated-ANP activation is an important hormonal mechanism in regulating adipose tissue function and body temperature upon cold exposure in mice.
Collapse
Affiliation(s)
- Xianrui Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
- MOH Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Wenguo Li
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
- MOH Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Tiantian Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
| | - Meng Liu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
- Correspondence: (Q.W.); (N.D.)
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China; (X.Z.); (W.L.); (T.Z.); (M.L.)
- MOH Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Correspondence: (Q.W.); (N.D.)
| |
Collapse
|
43
|
Natriuretic Peptide-Based Novel Therapeutics: Long Journeys of Drug Developments Optimized for Disease States. BIOLOGY 2022; 11:biology11060859. [PMID: 35741380 PMCID: PMC9219923 DOI: 10.3390/biology11060859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 12/11/2022]
Abstract
Simple Summary Natriuretic peptides are endogenous hormones produced in the heart and vascular endothelium, and they enable cardiorenal protective actions or bone growth via cGMP stimulation through their receptor guanylyl cyclase receptor A or B. To optimize the drug for each disease state, we must consider drug metabolism, delivery systems, and target receptor(s). This review summarizes attempts to develop novel natriuretic peptide-based therapeutics, including novel designer natriuretic peptides and oral drugs to enhance endogenous natriuretic peptides. We introduce some therapeutics that have been successful in clinical practice, as well as the prospective drug developments in the natriuretic peptide system for disease states. Abstract The field of natriuretic peptides (NPs) as an endocrine hormone has been developing since 1979. There are three peptides in humans: atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP), which bind to the guanylyl cyclase-A (GC-A) receptor (also called natriuretic peptide receptor-A (NPR-A)), and C-type natriuretic peptide (CNP), which binds to the GC-B receptor (also called the NPR-B) and then synthesizes intracellular cGMP. GC-A receptor stimulation has natriuretic, vasodilatory, cardiorenal protective and anti-renin–angiotensin–aldosterone system actions, and GC-B receptor stimulation can suppress myocardial fibrosis and can activate bone growth before epiphyseal plate closure. These physiological effects are useful as therapeutics for some disease states, such as heart failure, hypertension, and dwarfism. To optimize the therapeutics for each disease state, we must consider drug metabolism, delivery systems, and target receptor(s). We review the cardiac NP system; new designer NPs, such as modified/combined NPs and modified peptides that can bind to not only NP receptors but receptors for other systems; and oral drugs that enhance endogenous NP activity. Finally, we discuss prospective drug discoveries and the development of novel NP therapeutics.
Collapse
|
44
|
Ding K, Gui Y, Hou X, Ye L, Wang L. Transient Receptor Potential Channels, Natriuretic Peptides, and Angiotensin Receptor-Neprilysin Inhibitors in Patients With Heart Failure. Front Cardiovasc Med 2022; 9:904881. [PMID: 35722101 PMCID: PMC9204593 DOI: 10.3389/fcvm.2022.904881] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) remains the leading cause of death, morbidity, and medical expenses worldwide. Treatments for HF with reduced ejection fraction have progressed in recent years; however, acute decompensated heart failure remains difficult to treat. The transient receptor potential (TRP) channel family plays roles in various cardiovascular diseases, responding to neurohormonal and mechanical load stimulation. Thus, TRP channels are promising targets for drug discovery, and many studies have evaluated the roles of TRP channels expressed on pain neurons. The natriuretic peptide (NP) family of proteins regulates blood volume, natriuresis, and vasodilation and can antagonize the renin-angiotensin-aldosterone system and participate in the pathogenesis of major cardiovascular diseases, such as HF, coronary atherosclerotic heart disease, and left ventricular hypertrophy. NPs are degraded by neprilysin, and the blood level of NPs has predictive value in the diagnosis and prognostic stratification of HF. In this review, we discuss the relationships between typical TRP family channels (e.g., transient receptor potential cation channel subfamily V member 1 andTRPV1, transient receptor potential cation channel subfamily C member 6) and the NP system (e.g., atrial NP, B-type NP, and C-type NP) and their respective roles in HF. We also discuss novel drugs introduced for the treatment of HF.
Collapse
Affiliation(s)
- Kun Ding
- Bengbu Medical College, Bengbu, China
- Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Yang Gui
- Bengbu Medical College, Bengbu, China
- Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Xu Hou
- Bengbu Medical College, Bengbu, China
- Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Lifang Ye
- Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Lihong Wang
- Zhejiang Provincial People’s Hospital, Hangzhou, China
| |
Collapse
|
45
|
Itoh H, Tanaka M. “Greedy Organs Hypothesis” for sugar and salt in the pathophysiology of non-communicable diseases in relation to sodium-glucose co-transporters in the intestines and the kidney. Metabol Open 2022; 13:100169. [PMID: 35198947 PMCID: PMC8844901 DOI: 10.1016/j.metop.2022.100169] [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: 01/26/2022] [Revised: 02/07/2022] [Accepted: 02/07/2022] [Indexed: 11/05/2022] Open
Abstract
Deposition of visceral fat and insulin resistance play central role in the development of non-communicable diseases (NCDs) including obesity, hypertension and type 2 diabetes. However, we shed more light upon the intestines and the kidney as a strong driver of NCDs. Based upon unexpected outcomes of clinical trials using sodium-glucose cotransporter (SGLT) 2 inhibitors to demonstrate their actions for not only body weight reduction and blood glucose fall but also remarkable cardiorenal protection, we speculate that hyperfunction of the intestines and the kidney is one of critical contributing factors for initiation of NCDs. By detecting high amount of glucose and sodium chloride around them by sweet/salt taste sensors, the intestines and the kidney are designed to (re)absorb these nutrients by up-regulating SGLT1 or SGLT2. We designate these hyperfunctioning organs for nutrient uptake as “greedy organs”. The greedy organs can induce NCDs (“greedy organ hypothesis”). SGLTs are regulated by glucose and sodium chloride, and SGLTs or other genes can be “greedy genes.” Regulating factors for greedy organs are renin-angiotensin system, renal sympathetic nervous activity, gut inflammation/microbiota or oxidative stress. Mitigation of organ greediness by SGLT2 inhibitors, ketone bodies, bariatric surgery, and regular lifestyle to keep rhythmicity of biological clock are promising. We propose the concept of “Greedy Organs” hypothesis as a possible cause of NCDs. Clinical implication of greedy kidney is supported by the effect of SGLT2 inhibitors. The significance of greedy intestines is suggested by the effect of bariatric surgery. The intestines and kidney become hyperactive through upregulation of SGLT1 or 2. To mitigate “greedy organs” should be a promising strategy against NCDs.
Collapse
|
46
|
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
|
47
|
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
|
48
|
Rukavina Mikusic NL, Kouyoumdzian NM, Puyó AM, Fernández BE, Choi MR. Role of natriuretic peptides in the cardiovascular-adipose communication: a tale of two organs. Pflugers Arch 2022; 474:5-19. [PMID: 34173888 DOI: 10.1007/s00424-021-02596-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 05/31/2021] [Accepted: 06/11/2021] [Indexed: 12/23/2022]
Abstract
Natriuretic peptides have long been known for their cardiovascular function. However, a growing body of evidence emphasizes the role of natriuretic peptides in the energy metabolism of several substrates in humans and animals, thus interrelating the heart, as an endocrine organ, with various insulin-sensitive tissues and organs such as adipose tissue, muscle skeletal, and liver. Adipose tissue dysfunction is associated with altered regulation of the natriuretic peptide system, also indicated as a natriuretic disability. Evidence points to a contribution of this natriuretic disability to the development of obesity, type 2 diabetes mellitus, and cardiometabolic complications; although the causal relationship is not fully understood at present. However, targeting the natriuretic peptide pathway may improve metabolic health in obesity and type 2 diabetes mellitus. This review will focus on the current literature on the metabolic functions of natriuretic peptides with emphasis on lipid metabolism and insulin sensitivity. Natriuretic peptide system alterations could be proposed as one of the linking mechanisms between adipose tissue dysfunction and cardiovascular disease.
Collapse
Affiliation(s)
- Natalia Lucía Rukavina Mikusic
- Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Nicolás Martín Kouyoumdzian
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana María Puyó
- Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Marcelo Roberto Choi
- Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto Universitario de Ciencias de la Salud, Fundación H.A. Barceló, Buenos Aires, Argentina
| |
Collapse
|
49
|
King NE, Brittain E. Emerging therapies: The potential roles SGLT2 inhibitors, GLP1 agonists, and ARNI therapy for ARNI pulmonary hypertension. Pulm Circ 2022; 12:e12028. [PMID: 35506082 PMCID: PMC9052991 DOI: 10.1002/pul2.12028] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a highly morbid condition. PH due to left heart disease (PH-LHD) has no specific therapies and pulmonary arterial hypertension (PAH) has substantial residual risk despite several approved therapies. Multiple lines of experimental evidence link metabolic dysfunction to the pathogenesis and outcomes in PH-LHD and PAH, and novel metabolic agents hold promise to improve outcomes in these populations. The antidiabetic sodium-glucose cotransporter 2 (SGLT2) inhibitors and glucagon-like peptide-1 (GLP1) agonists targeting metabolic dysfunction and improve outcomes in patients with LHD but have not been tested specifically in patients with PH. The angiotensin receptor/neprilysin inhibitors (ARNIs) produce significant improvements in cardiac hemodynamics and may improve metabolic dysfunction that could benefit the pulmonary circulation and right ventricle function. On the basis of promising preclinical work with these medications and clinical rationale, we explore the potential of SGLT2 inhibitors, GLP1 agonists, and ARNIs as therapies for both PH-LHD and PAH.
Collapse
Affiliation(s)
| | - Evan Brittain
- Department of Medicine, Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
50
|
Moro C. [Natriuretic peptides control of skeletal muscle oxidative metabolism]. Med Sci (Paris) 2021; 37 Hors série n° 1:19-21. [PMID: 34878388 DOI: 10.1051/medsci/2021185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Besides their well-known effect in the regulation of blood volume, natriuretic peptides have emerged during the last two decades as key metabolic hormones linking cardiac function to energy metabolism. Recent work from our laboratory underscores a new role of these peptides in the control of oxidative metabolism in skeletal muscle and open novel perspectives in the field of chronic diseases affecting skeletal muscles.
Collapse
Affiliation(s)
- Cédric Moro
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Équipe MetaDiab, Inserm/Université Paul Sabatier UMR1297, CHU Rangueil, 1 avenue Jean Poulhès, 31432 Toulouse Cedex 4, France
| |
Collapse
|