1
|
Li H, Sun S, Guo W, Wang L, Zhang Z, Zhang Y, Zhang C, Liu M, Zhang S, Niu Y, Dong N, Wu Q. Positively charged cytoplasmic residues in corin prevent signal peptidase cleavage and endoplasmic reticulum retention. Commun Biol 2025; 8:89. [PMID: 39833422 PMCID: PMC11756421 DOI: 10.1038/s42003-025-07545-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 01/14/2025] [Indexed: 01/30/2025] Open
Abstract
Positively charged residues are commonly located near the cytoplasm-membrane interface, which is known as the positive-inside rule in membrane topology. The mechanism underlying the function of these charged residues remains poorly understood. Herein, we studied the function of cytoplasmic residues in corin, a type II transmembrane serine protease in cardiovascular biology. We found that the positively charged residue at the cytoplasm-membrane interface of corin was not a primary determinant in membrane topology but probably served as a charge-repulsion mechanism in the endoplasmic reticulum (ER) to prevent interactions with proteins in the ER, including the signal peptidase. Substitution of the positively charged residue with a neutral or acidic residue resulted in corin secretion likely due to signal peptidase cleavage. In signal peptidase-deficient cells, the mutant corin proteins were not secreted but retained in the ER. Similar results were found in the low-density lipoprotein receptor and matriptase-2 that have positively charged residues at and near the cytoplasm-membrane interface. These results provide important insights into the role of the positively charged cytoplasmic residues in mammalian single-pass transmembrane proteins.
Collapse
Affiliation(s)
- Hui Li
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Suzhou Medical College, Soochow University, Suzhou, 215123, China
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Shijin Sun
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Suzhou Medical College, Soochow University, Suzhou, 215123, China
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Wenjun Guo
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Suzhou Medical College, Soochow University, Suzhou, 215123, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lina Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Suzhou Medical College, Soochow University, Suzhou, 215123, China
| | - Zihao Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Suzhou Medical College, Soochow University, Suzhou, 215123, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yue Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Suzhou Medical College, Soochow University, Suzhou, 215123, China
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Ce Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Suzhou Medical College, Soochow University, Suzhou, 215123, China
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Meng Liu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Suzhou Medical College, Soochow University, Suzhou, 215123, China
| | - Shengnan Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Suzhou Medical College, Soochow University, Suzhou, 215123, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yayan Niu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Suzhou Medical College, Soochow University, Suzhou, 215123, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Suzhou Medical College, Soochow University, Suzhou, 215123, China.
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Suzhou Medical College, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
2
|
Pinheiro F, Lail H, Neves JS, Negrão R, Wanders D. Sulfur Amino Acid Restriction Mitigates High-Fat Diet-Induced Molecular Alterations in Cardiac Remodeling Primarily via FGF21-Independent Mechanisms. Nutrients 2024; 16:4347. [PMID: 39770968 PMCID: PMC11677450 DOI: 10.3390/nu16244347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Dietary sulfur amino acid restriction (SAAR) elicits various health benefits, some mediated by fibroblast growth factor 21 (FGF21). However, research on SAAR's effects on the heart is limited and presents mixed findings. This study aimed to evaluate SAAR-induced molecular alterations associated with cardiac remodeling and their dependence on FGF21. Methods: Male C57BL/6J wild-type and FGF21 knockout mice were randomized into four dietary regimens, including normal fat and high-fat diets (HFDs) with and without SAAR, over five weeks. Results: SAAR significantly reduced body weight and visceral adiposity while increasing serum FGF21 levels. In the heart, SAAR-induced molecular metabolic alterations are indicative of enhanced lipid utilization, glucose uptake, and mitochondrial biogenesis. SAAR also elicited opposing effects on the cardiac gene expression of FGF21 and adiponectin. Regarding cellular stress responses, SAAR mitigated the HFD-induced increase in the cardiac expression of genes involved in oxidative stress, inflammation, and apoptosis, while upregulating antioxidative genes. Structurally, SAAR did not induce alterations indicative of cardiac hypertrophy and it counteracted HFD-induced fibrotic gene expression. Overall, most alterations induced by SAAR were FGF21-independent, except for those related to lipid utilization and glucose uptake. Conclusions: Altogether, SAAR promotes cardiac alterations indicative of physiological rather than pathological remodeling, primarily through FGF21-independent mechanisms.
Collapse
Affiliation(s)
- Filipe Pinheiro
- Department of Nutrition, Georgia State University, 140 Decatur St SE, Atlanta, GA 30303, USA; (F.P.); (H.L.)
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Hannah Lail
- Department of Nutrition, Georgia State University, 140 Decatur St SE, Atlanta, GA 30303, USA; (F.P.); (H.L.)
- Department of Chemistry, Georgia State University, 100 Piedmont Ave., Atlanta, GA 30303, USA
| | - João Sérgio Neves
- Department of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário de São João, Alameda Hernâni Monteiro, 4200-319 Porto, Portugal;
- Unit of Cardiovascular Research and Development—Unic@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Rita Negrão
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Center for Health Technology and Services Research—CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Desiree Wanders
- Department of Nutrition, Georgia State University, 140 Decatur St SE, Atlanta, GA 30303, USA; (F.P.); (H.L.)
| |
Collapse
|
3
|
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
|
4
|
Li W, Zhang X, Zhou Z, Guo W, Wang M, Zhou T, Liu M, Wu Q, Dong N. Cardiac corin and atrial natriuretic peptide regulate liver glycogen metabolism and glucose homeostasis. Cardiovasc Diabetol 2024; 23:383. [PMID: 39468553 PMCID: PMC11520433 DOI: 10.1186/s12933-024-02475-w] [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: 08/02/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Cardiovascular function and metabolic homeostasis are closely linked, but the underlying mechanisms are not fully understood. Corin is a protease that activates atrial natriuretic peptide (ANP), an essential hormone for normal blood pressure and cardiac function. The goal of this study is to investigate a potential corin and ANP function in regulating liver glycogen metabolism and glucose homeostasis. METHODS Liver glycogen and blood glucose levels were analyzed in Corin or Nppa (encoding ANP) knockout (KO) mice. ANP signaling was examined in livers from Corin and Nppa KO mice and in cultured human and mouse hepatocytes by western blotting. RESULTS We found that Corin and Nppa KO mice had reduced liver glycogen contents and increased blood glucose levels. By analyzing conditional KO mice lacking either cardiac or renal Corin, we showed that cardiac corin and ANP act in an endocrine manner to enhance cGMP-protein kinase G (PKG)-AKT-GSK3 signaling in hepatocytes. In cultured hepatocytes, ANP treatment stimulated PKG signaling, glucose uptake, and glycogen production, which could be blocked by small molecule PKG and AKT inhibitors. CONCLUSIONS Our results indicate that corin and ANP are important regulators in liver glycogen metabolism and glucose homeostasis, suggesting that defects in the corin and ANP pathway may contribute to both cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Wenguo Li
- 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
| | - Xianrui Zhang
- 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
| | - Zibin Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenjun Guo
- 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
| | - Mengting Wang
- 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
| | - Tiantian Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Meng Liu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China.
| | - 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.
| |
Collapse
|
5
|
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
|
6
|
Ding Y, Su J, Shan B, Fu X, Zheng G, Wang J, Wu L, Wang F, Chai X, Sun H, Zhang J. Brown adipose tissue-derived FGF21 mediates the cardioprotection of dexmedetomidine in myocardial ischemia/reperfusion injury. Sci Rep 2024; 14:18292. [PMID: 39112671 PMCID: PMC11306229 DOI: 10.1038/s41598-024-69356-w] [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: 05/16/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024] Open
Abstract
Brown adipose tissue (BAT) plays a critical role in regulating cardiovascular homeostasis through the secretion of adipokines, such as fibroblast growth factor 21 (FGF21). Dexmedetomidine (DEX) is a selective α2-adrenergic receptor agonist with a protection against myocardial ischemia/reperfusion injury (MI/RI). It remains largely unknown whether or not BAT-derived FGF21 is involved in DEX-induced cardioprotection in the context of MI/RI. Herein, we demonstrated that DEX alleviated MI/RI and improved heart function through promoting the release of FGF21 from interscapular BAT (iBAT). Surgical iBAT depletion or supplementation with a FGF21 neutralizing antibody attenuated the beneficial effects of DEX. AMPK/PGC1α signaling-induced fibroblast growth factor 21 (FGF21) release in brown adipocytes is required for DEX-mediated cardioprotection since blockade of the AMPK/PGC1α axis weakened the salutary effects of DEX. Co-culture experiments showed that DEX-induced FGF21 from brown adipocytes increased the resistance of cardiomyocytes to hypoxia/reoxygenation (H/R) injury via modulating the Keap1/Nrf2 pathway. Our results provided robust evidence that the BAT-cardiomyocyte interaction is required for DEX cardioprotection, and revealed an endocrine role of BAT in DEX-mediating protection of hearts against MIRI.
Collapse
Affiliation(s)
- Yi Ding
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China
| | - Jiabao Su
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China
| | - Beiying Shan
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China
| | - Xiao Fu
- Laboratory of Metabolic and Inflammatory Diseases, Wuxi School of Medicine, Jiangnan University, No.1800, Lihu Road, Wuxi, 214125, People's Republic of China
| | - Guanli Zheng
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China
| | - Jiwen Wang
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China
| | - Lixue Wu
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China
| | - Fangming Wang
- Department of Rheumatology and Immunology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, 214125, People's Republic of China
| | - Xiaoying Chai
- Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, 214125, People's Republic of China
| | - Haijian Sun
- Laboratory of Metabolic and Inflammatory Diseases, Wuxi School of Medicine, Jiangnan University, No.1800, Lihu Road, Wuxi, 214125, People's Republic of China.
| | - Jiru Zhang
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214125, People's Republic of China.
| |
Collapse
|
7
|
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
|
8
|
Gu X, Liu M, Wang M, Wang K, Zhou T, Wu Q, Dong N. Corin deficiency alleviates mucosal lesions in a mouse model of colitis induced by dextran sulfate sodium. Life Sci 2024; 339:122446. [PMID: 38246520 DOI: 10.1016/j.lfs.2024.122446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024]
Abstract
AIMS High dietary salt consumption is a risk factor for inflammatory bowel disease (IBD). Corin is a protease that activates atrial natriuretic peptide (ANP), thereby regulating sodium homeostasis. Corin acts in multiple tissues, including the intestine. In mice, corin deficiency impairs intestinal sodium excretion. This study aims to examine if reduced intestinal sodium excretion alters the pathophysiology of IBD. MAIN METHODS Wild-type (WT), Corin knockout (KO), and Corin kidney conditional KO (kcKO) mice were tested in a colitis model induced by dextran sulfide sodium (DSS). Effects of ANP on DSS-induced colitis were tested in WT and Corin KO mice. Body weight changes in the mice were monitored. Necropsy, histological analysis, and immunostaining studies were conducted to examine colon length and mucosal lesions. Fecal sodium levels were measured. RT-PCR was done to analyze proinflammatory genes in colon samples. KEY FINDINGS DSS-treated Corin KO mice had an ameliorated colitis phenotype with less body weight loss, longer colon lengths, smaller mucosal lesions, lower disease scores, more preserved goblet cells, and suppressed proinflammatory genes in the colon. In longitudinal studies, the DSS-treated Corin KO mice had delayed onset of colon mucosal lesions. ANP administration lessened the colitis in WT, but not Corin KO, mice. Analyses of WT, Corin KO, and Corin kcKO mice indicated that fecal sodium excretion, controlled by intestinal corin, may regulate inflammatory responses in DSS-induced colitis in mice. SIGNIFICANCE Our findings indicate a role of corin in intestinal pathophysiology, suggesting that reduced intestinal sodium level may offer protective benefits against IBD.
Collapse
Affiliation(s)
- Xiabing Gu
- Jiangsu Institute of Hematology, The First Affiliated Hospital and 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
| | - Meng Liu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Mengting Wang
- Jiangsu Institute of Hematology, The First Affiliated Hospital and 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
| | - Kun Wang
- Jiangsu Institute of Hematology, The First Affiliated Hospital and 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
| | - Tiantian Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China.
| | - Ningzheng Dong
- Jiangsu Institute of Hematology, The First Affiliated Hospital and 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.
| |
Collapse
|
9
|
Ceddia RP, Zurawski Z, Thompson Gray A, Adegboye F, McDonald-Boyer A, Shi F, Liu D, Maldonado J, Feng J, Li Y, Alford S, Ayala JE, McGuinness OP, Collins S, Hamm HE. Gβγ-SNAP25 exocytotic brake removal enhances insulin action, promotes adipocyte browning, and protects against diet-induced obesity. J Clin Invest 2023; 133:e160617. [PMID: 37561580 PMCID: PMC10541194 DOI: 10.1172/jci160617] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/08/2023] [Indexed: 08/12/2023] Open
Abstract
Negative regulation of exocytosis from secretory cells is accomplished through inhibitory signals from Gi/o GPCRs by Gβγ subunit inhibition of 2 mechanisms: decreased calcium entry and direct interaction of Gβγ with soluble N-ethylmaleimide-sensitive factor attachment protein (SNAP) receptor (SNARE) plasma membrane fusion machinery. Previously, we disabled the second mechanism with a SNAP25 truncation (SNAP25Δ3) that decreased Gβγ affinity for the SNARE complex, leaving exocytotic fusion and modulation of calcium entry intact and removing GPCR-Gβγ inhibition of SNARE-mediated exocytosis. Here, we report substantial metabolic benefit in mice carrying this mutation. Snap25Δ3/Δ3 mice exhibited enhanced insulin sensitivity and beiging of white fat. Metabolic protection was amplified in Snap25Δ3/Δ3 mice challenged with a high-fat diet. Glucose homeostasis, whole-body insulin action, and insulin-mediated glucose uptake into white adipose tissue were improved along with resistance to diet-induced obesity. Metabolic protection in Snap25Δ3/Δ3 mice occurred without compromising the physiological response to fasting or cold. All metabolic phenotypes were reversed at thermoneutrality, suggesting that basal autonomic activity was required. Direct electrode stimulation of sympathetic neuron exocytosis from Snap25Δ3/Δ3 inguinal adipose depots resulted in enhanced and prolonged norepinephrine release. Thus, the Gβγ-SNARE interaction represents a cellular mechanism that deserves further exploration as an additional avenue for combating metabolic disease.
Collapse
Affiliation(s)
- Ryan P. Ceddia
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Zack Zurawski
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | - Feyisayo Adegboye
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Fubiao Shi
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Dianxin Liu
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jose Maldonado
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Julio E. Ayala
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Owen P. McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Sheila Collins
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Heidi E. Hamm
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
10
|
Wu Q. Natriuretic Peptide Signaling in Uterine Biology and Preeclampsia. Int J Mol Sci 2023; 24:12309. [PMID: 37569683 PMCID: PMC10418983 DOI: 10.3390/ijms241512309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Endometrial decidualization is a uterine process essential for spiral artery remodeling, embryo implantation, and trophoblast invasion. Defects in endometrial decidualization and spiral artery remodeling are important contributing factors in preeclampsia, a major disorder in pregnancy. Atrial natriuretic peptide (ANP) is a cardiac hormone that regulates blood volume and pressure. ANP is also generated in non-cardiac tissues, such as the uterus and placenta. In recent human genome-wide association studies, multiple loci with genes involved in natriuretic peptide signaling are associated with gestational hypertension and preeclampsia. In cellular experiments and mouse models, uterine ANP has been shown to stimulate endometrial decidualization, increase TNF-related apoptosis-inducing ligand expression and secretion, and enhance apoptosis in arterial smooth muscle cells and endothelial cells. In placental trophoblasts, ANP stimulates adenosine 5'-monophosphate-activated protein kinase and the mammalian target of rapamycin complex 1 signaling, leading to autophagy inhibition and protein kinase N3 upregulation, thereby increasing trophoblast invasiveness. ANP deficiency impairs endometrial decidualization and spiral artery remodeling, causing a preeclampsia-like phenotype in mice. These findings indicate the importance of natriuretic peptide signaling in pregnancy. This review discusses the role of ANP in uterine biology and potential implications of impaired ANP signaling in preeclampsia.
Collapse
Affiliation(s)
- Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
| |
Collapse
|
11
|
Wu Q, Li S, Zhang X, Dong N. Type II Transmembrane Serine Proteases as Modulators in Adipose Tissue Phenotype and Function. Biomedicines 2023; 11:1794. [PMID: 37509434 PMCID: PMC10376093 DOI: 10.3390/biomedicines11071794] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Adipose tissue is a crucial organ in energy metabolism and thermoregulation. Adipose tissue phenotype is controlled by various signaling mechanisms under pathophysiological conditions. Type II transmembrane serine proteases (TTSPs) are a group of trypsin-like enzymes anchoring on the cell surface. These proteases act in diverse tissues to regulate physiological processes, such as food digestion, salt-water balance, iron metabolism, epithelial integrity, and auditory nerve development. More recently, several members of the TTSP family, namely, hepsin, matriptase-2, and corin, have been shown to play a role in regulating lipid metabolism, adipose tissue phenotype, and thermogenesis, via direct growth factor activation or indirect hormonal mechanisms. In mice, hepsin deficiency increases adipose browning and protects from high-fat diet-induced hyperglycemia, hyperlipidemia, and obesity. Similarly, matriptase-2 deficiency increases fat lipolysis and reduces obesity and hepatic steatosis in high-fat diet-fed mice. In contrast, corin deficiency increases white adipose weights and cell sizes, suppresses adipocyte browning and thermogenic responses, and causes cold intolerance in mice. These findings highlight an important role of TTSPs in modifying cellular phenotype and function in adipose tissue. In this review, we provide a brief description about TTSPs and discuss recent findings regarding the role of hepsin, matriptase-2, and corin in regulating adipose tissue phenotype, energy metabolism, and thermogenic responses.
Collapse
Affiliation(s)
- Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
| | - Shuo Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xianrui Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, Soochow University, Suzhou 215006, China
| |
Collapse
|
12
|
Wu Q, Burley G, Li L, Lin S, Shi Y. The role of dietary salt in metabolism and energy balance: Insights beyond cardiovascular disease. Diabetes Obes Metab 2023; 25:1147-1161. [PMID: 36655379 PMCID: PMC10946535 DOI: 10.1111/dom.14980] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/20/2023]
Abstract
Dietary salt (NaCl) is essential to an organism's survival. However, today's diets are dominated by excessive salt intake, which significantly impacts individual and population health. High salt intake is closely linked to cardiovascular disease (CVD), especially hypertension, through a number of well-studied mechanisms. Emerging evidence indicates that salt overconsumption may also be associated with metabolic disorders. In this review, we first summarize recent updates on the mechanisms of salt-induced CVD, the effects of salt reduction and the use of salt substitution as a therapy. Next, we focus on how high salt intake can impact metabolism and energy balance, describing the mechanisms through which this occurs, including leptin resistance, the overproduction of fructose and ghrelin, insulin resistance and altered hormonal factors. A further influence on metabolism worth noting is the reported role of salt in inducing thermogenesis and increasing body temperature, leading to an increase in energy expenditure. While this result could be viewed as a positive metabolic effect because it promotes a negative energy balance to combat obesity, caution must be taken with this frame of thinking given the deleterious consequences of chronic high salt intake on cardiovascular health. Nevertheless, this review highlights the importance of salt as a noncaloric nutrient in regulating whole-body energy homeostasis. Through this review, we hope to provide a scientific framework for future studies to systematically address the metabolic impacts of dietary salt and salt replacement treatments. In addition, we hope to form a foundation for future clinical trials to explore how these salt-induced metabolic changes impact obesity development and progression, and to elucidate the regulatory mechanisms that drive these changes, with the aim of developing novel therapeutics for obesity and CVD.
Collapse
Affiliation(s)
- Qi Wu
- Obesity and Metabolic Disease Research GroupGarvan Institute of Medical ResearchSydneyNew South WalesAustralia
- Centre of Neurological and Metabolic Researchthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - George Burley
- Obesity and Metabolic Disease Research GroupGarvan Institute of Medical ResearchSydneyNew South WalesAustralia
| | - Li‐Cheng Li
- Centre of Neurological and Metabolic Researchthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - Shu Lin
- Obesity and Metabolic Disease Research GroupGarvan Institute of Medical ResearchSydneyNew South WalesAustralia
- Centre of Neurological and Metabolic Researchthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
| | - Yan‐Chuan Shi
- Obesity and Metabolic Disease Research GroupGarvan Institute of Medical ResearchSydneyNew South WalesAustralia
- Centre of Neurological and Metabolic Researchthe Second Affiliated Hospital of Fujian Medical UniversityQuanzhouChina
- School of Clinical Medicine, St Vincent's Clinical CampusFaculty of Medicine and HealthSydneyNew South WalesAustralia
| |
Collapse
|
13
|
Couch CA, Fowler LA, Parcha V, Arora P, Gower BA. Associations of atrial natriuretic peptide with measures of insulin and adipose depots. Physiol Rep 2023; 11:e15625. [PMID: 36905117 PMCID: PMC10006508 DOI: 10.14814/phy2.15625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/06/2023] [Accepted: 02/12/2023] [Indexed: 03/12/2023] Open
Abstract
Low concentrations of natriuretic peptides (NPs) have been associated with greater risk for Type 2 diabetes (T2D). African American individuals (AA) have lower NP levels and are disproportionately burdened by T2D. The purpose of this study was to test the hypothesis that higher post-challenge insulin in AA adults is associated with lower plasma N-terminal pro-atrial natriuretic peptide (NT-proANP). A secondary purpose was to explore associations between NT-proANP and adipose depots. Participants were 112 AA and European American (EA) adult men and women. Measures of insulin were obtained from an oral glucose tolerance test and hyperinsulinemic-euglycemic glucose clamp. Total and regional adipose depots were measured from DXA and MRI. Multiple linear regression analysis was used to assess associations of NT-proANP with measures of insulin and adipose depots. Lower NT-proANP concentrations in AA participants was not independent of 30-min insulin area under the curve (AUC). NT-proANP was inversely associated with 30-min insulin AUC in AA participants, and with fasting insulin and HOMA-IR in EA participants. Thigh subcutaneous adipose tissue and perimuscular adipose tissue were positively associated with NT-proANP in EA participants. Higher post-challenge insulin may contribute to lower ANP concentrations in AA adults.
Collapse
Affiliation(s)
- Catharine A. Couch
- Department of Nutrition SciencesUniversity of Alabama at BirminghamBirminghamALUSA
| | - Lauren A. Fowler
- Department of Nutrition SciencesUniversity of Alabama at BirminghamBirminghamALUSA
| | - Vibhu Parcha
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamALUSA
| | - Pankaj Arora
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamALUSA
| | - Barbara A. Gower
- Department of Nutrition SciencesUniversity of Alabama at BirminghamBirminghamALUSA
| |
Collapse
|
14
|
Wang B, Zhao M, Su Z, Jin B, Yang X, Zhang C, Guo B, Li J, Hong W, Liu J, Zhao Y, Hou Y, Lai F, Zhang W, Qin L, Zhang W, Luo J, Zheng R. RIIβ-PKA in GABAergic Neurons of Dorsal Median Hypothalamus Governs White Adipose Browning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205173. [PMID: 36529950 PMCID: PMC9929258 DOI: 10.1002/advs.202205173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/18/2022] [Indexed: 06/17/2023]
Abstract
The RIIβ subunit of cAMP-dependent protein kinase A (PKA) is expressed in the brain and adipose tissue. RIIβ-knockout mice show leanness and increased UCP1 in brown adipose tissue. The authors have previously reported that RIIβ reexpression in hypothalamic GABAergic neurons rescues the leanness. However, whether white adipose tissue (WAT) browning contributes to the leanness and whether RIIβ-PKA in these neurons governs WAT browning are unknown. Here, this work reports that RIIβ-KO mice exhibit a robust WAT browning. RIIβ reexpression in dorsal median hypothalamic GABAergic neurons (DMH GABAergic neurons) abrogates WAT browning. Single-cell sequencing, transcriptome sequencing, and electrophysiological studies show increased GABAergic activity in DMH GABAergic neurons of RIIβ-KO mice. Activation of DMH GABAergic neurons or inhibition of PKA in these neurons elicits WAT browning and thus lowers body weight. These findings reveal that RIIβ-PKA in DMH GABAergic neurons regulates WAT browning. Targeting RIIβ-PKA in DMH GABAergic neurons may offer a clinically useful way to promote WAT browning for treating obesity and other metabolic disorders.
Collapse
Affiliation(s)
- Bingwei Wang
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Miao Zhao
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Zhijie Su
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Baohua Jin
- Department of PharmacologyInstitution of Chinese Integrative MedicineHebei Medical UniversityShijiazhuang050017P. R. China
| | - Xiaoning Yang
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Chenyu Zhang
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Bingbing Guo
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Jiebo Li
- Institute of Medical PhotonicsBeijing Advanced Innovation Center for Biomedical EngineeringSchool of Biological Science and Medical EngineeringBeihang UniversityBeijing100191P. R. China
| | - Weili Hong
- Institute of Medical PhotonicsBeijing Advanced Innovation Center for Biomedical EngineeringSchool of Biological Science and Medical EngineeringBeihang UniversityBeijing100191P. R. China
| | - Jiarui Liu
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Yun Zhao
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Yujia Hou
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Futing Lai
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Wei Zhang
- Department of PharmacologyInstitution of Chinese Integrative MedicineHebei Medical UniversityShijiazhuang050017P. R. China
| | - Lihua Qin
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Weiguang Zhang
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Jianyuan Luo
- Department of Medical GeneticsSchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Ruimao Zheng
- Department of AnatomyHistology and EmbryologySchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
- Neuroscience Research InstituteKey Laboratory for Neuroscience of Ministry of EducationKey Laboratory for Neuroscience of National Health Commission of the People's Republic of ChinaPeking UniversityBeijing100191P. R. China
| |
Collapse
|
15
|
Lu G, Hu R, Tao T, Hu M, Dong Z, Wang C. Regulatory role of atrial natriuretic peptide in brown adipose tissue: A narrative review. Obes Rev 2023; 24:e13522. [PMID: 36336901 DOI: 10.1111/obr.13522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/15/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022]
Abstract
Atrial natriuretic peptide (ANP) has been considered to exert an essential role as a cardiac secretory hormone in the regulation of hemodynamic homeostasis. As the research progresses, the role of ANP in the crosstalk between heart and lipid metabolism has become an interesting topic that is attracting the interest of researchers. The regulation of ANP in lipid metabolism shows favorable effects, particularly the activation of brown adipose tissue (BAT). The complex regulatory network of ANP on BAT has not been fully outlined. This narrative review critically evaluated the existing literature on the regulatory effects of ANP on BAT. In general, we have summarized the expression of ANP and its receptors in various human tissues, analyzed the progress of research on the relationship between the ANP and BAT, and described several potential pathways of ANP to BAT. Exogenous ANP, natriuretic peptide receptor C (NPRC) deficiency, cold exposure, bariatric surgery, and cardiac or renal insufficiency could all contribute to BAT expression by increasing circulating ANP levels.
Collapse
Affiliation(s)
- Guanhua Lu
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| | - Ruixiang Hu
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| | - Tian Tao
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| | - Min Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Zhiyong Dong
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, The University of Hong Kong and Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
16
|
Moxonidine ameliorates cardiac injury in rats with metabolic syndrome by regulating autophagy. Life Sci 2022; 312:121210. [PMID: 36410408 DOI: 10.1016/j.lfs.2022.121210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022]
Abstract
AIMS Reduced cardiac autophagy, ischemic injury, sympathetic overactivity, and apoptosis all contribute to metabolic syndrome (MetS)-associated cardiovascular risks. NR4A2, an orphan nuclear receptor NR4A family member, induces autophagy while suppressing apoptosis in myocardial infarction. Moxonidine, a sympathoinhibitor imidazoline1 receptor (I1R) agonist, has beneficial metabolic and hemodynamic effects; however, whether autophagy and/or NR4A2 signaling are involved in moxonidine's cardiovascular effects via I1R activation, is unknown, and is the aim of this study. MATERIALS AND METHODS To induce MetS, rats were fed 3 % salt in their diet and 10 % fructose in their drinking water for 12 weeks. MetS-rats were given either moxonidine (6 mg/kg/day, gavage), efaroxan (I1R antagonist, 0.6 mg/kg/day, i.p), both treatments, or vehicles for the last two weeks. Blood pressure, lipid profile, and glycemic control were evaluated. Histopathological examination, circulating cardiac troponin I (c-TnI), proinflammatory interleukin-6 (IL-6), apoptosis (active caspase-3 and Fas-immunostaining), interstitial fibrosis [transforming growth factor-β1 (TGF-β1), Mallory's trichrome staining], and extracellular matrix remodeling [matrix metalloproteinase-9 (MMP-9)], were used to assess cardiac pathology. Cardiac NR4A2 and its downstream factor, p53, as well as autophagic flux markers, SQSTM1/p62, LC3, and Beclin-1 were also determined. KEY FINDINGS Moxonidine significantly ameliorated MetS-induced metabolic and hemodynamic derangements and the associated cardiac pathology. Moxonidine restored NR4A2 and p53 myocardial levels and enhanced autophagic flux via modulating SQSTM1/p62, LC3, and Beclin-1. Efaroxan reversed the majority of the moxonidine-induced improvements. SIGNIFICANCE The current study suggests that autophagy modulation via I1R activation is involved in moxonidine-mediated cardiac beneficial effects in MetS.
Collapse
|
17
|
Man W, Song X, Xiong Z, Gu J, Lin J, Gu X, Yu D, Li C, Jiang M, Zhang X, Yang Z, Cao Y, Zhang Y, Shu X, Wu D, Wang H, Ji G, Sun D. Exosomes derived from pericardial adipose tissues attenuate cardiac remodeling following myocardial infarction by Adipsin-regulated iron homeostasis. Front Cardiovasc Med 2022; 9:1003282. [PMID: 36172581 PMCID: PMC9510661 DOI: 10.3389/fcvm.2022.1003282] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
As a vital adipokine, Adipsin is closely associated with cardiovascular risks. Nevertheless, its role in the onset and development of cardiovascular diseases remains elusive. This study was designed to examine the effect of Adipsin on survival, cardiac dysfunction and adverse remodeling in the face of myocardial infarction (MI) injury. In vitro experiments were conducted to evaluate the effects of Adipsin on cardiomyocyte function in the face of hypoxic challenge and the mechanisms involved. Our results showed that Adipsin dramatically altered expression of proteins associated with iron metabolism and ferroptosis. In vivo results demonstrated that Adipsin upregulated levels of Ferritin Heavy Chain (FTH) while downregulating that of Transferrin Receptor (TFRC) in peri-infarct regions 1 month following MI. Adipsin also relieved post-MI-associated lipid oxidative stress as evidenced by decreased expression of COX2 and increased GPX4 level. Co-immunoprecipitation and immunofluorescence imaging prove a direct interaction between Adipsin and IRP2. As expected, cardioprotection provided by Adipsin depends on the key molecule of IRP2. These findings revealed that Adipsin could be efficiently delivered to the heart by exosomes derived from pericardial adipose tissues. In addition, Adipsin interacted with IRP2 to protect cardiomyocytes against ferroptosis and maintain iron homeostasis. Therefore, Adipsin-overexpressed exosomes derived from pericardial adipose tissues may be a promising therapeutic strategy to prevent adverse cardiac remodeling following ischemic heart injury.
Collapse
Affiliation(s)
- Wanrong Man
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xinglong Song
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhenyu Xiong
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jing Gu
- School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaoming Gu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an, China
| | - Duan Yu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhi Yang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yang Cao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaofei Shu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Dexi Wu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Haichang Wang
- Heart Hospital, Xi’an International Medical Center, Xi’an, China
- Haichang Wang,
| | - Gang Ji
- Department of Gastrointestinal Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Gang Ji,
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Dongdong Sun,
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Manaserh IH, Bledzka KM, Junker A, Grondolsky J, Schumacher SM. A Cardiac Amino-Terminal GRK2 Peptide Inhibits Maladaptive Adipocyte Hypertrophy and Insulin Resistance During Diet-Induced Obesity. JACC Basic Transl Sci 2022; 7:563-579. [PMID: 35818501 PMCID: PMC9270572 DOI: 10.1016/j.jacbts.2022.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 12/04/2022]
Abstract
Heart disease remains the leading cause of death, and mortality rates positively correlate with the presence of obesity and diabetes. Despite the correlation between cardiac and metabolic dysregulation, the mechanistic pathway(s) of interorgan crosstalk still remain undefined. This study reveals that cardiac-restricted expression of an amino-terminal peptide of GRK2 (βARKnt) preserves systemic and cardiac insulin responsiveness, and protects against adipocyte maladaptive hypertrophy in a diet-induced obesity model. These data suggest a cardiac-driven mechanism to ameliorate maladaptive cardiac remodeling and improve systemic metabolic homeostasis that may lead to new treatment modalities for cardioprotection in obesity and obesity-related metabolic syndromes.
Collapse
Key Words
- AS160, Akt substrate of 160 kilodaltons
- BAT, brown adipose tissue
- GRK2
- GRK2, G protein-coupled receptor kinase 2
- HFD, high-fat diet
- HOMA-IR, homeostatic model assessment of insulin resistance
- NLC, nontransgenic littermate control
- NP, natriuretic peptide
- NPR, natriuretic peptide receptor
- RER, respiratory exchange ratio
- T2D, type II diabetes
- Tg, transgenic
- beiging
- cardioprotection
- gWAT, gonadal white adipose tissue
- mTOR, mechanistic target of rapamycin protein kinase
- metabolism
- obesity
- βARKct, cardiac restricted expression of C-terminus domain of GRK2
- βARKnt, cardiac-restricted expression of N-terminus domain of GRK2
Collapse
Affiliation(s)
- Iyad H. Manaserh
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kamila M. Bledzka
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Alex Junker
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jessica Grondolsky
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sarah M. Schumacher
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
21
|
Molecular Mechanism of Induction of Bone Growth by the C-Type Natriuretic Peptide. Int J Mol Sci 2022; 23:ijms23115916. [PMID: 35682595 PMCID: PMC9180634 DOI: 10.3390/ijms23115916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/17/2022] [Accepted: 05/21/2022] [Indexed: 12/10/2022] Open
Abstract
The skeletal development process in the body occurs through sequential cellular and molecular processes called endochondral ossification. Endochondral ossification occurs in the growth plate where chondrocytes differentiate from resting, proliferative, hypertrophic to calcified zones. Natriuretic peptides (NPTs) are peptide hormones with multiple functions, including regulation of blood pressure, water-mineral balance, and many metabolic processes. NPTs secreted from the heart activate different tissues and organs, working in a paracrine or autocrine manner. One of the natriuretic peptides, C-type natriuretic peptide-, induces bone growth through several mechanisms. This review will summarize the knowledge, including the newest discoveries, of the mechanism of CNP activation in bone growth.
Collapse
|
22
|
Niewiadomska J, Gajek-Marecka A, Gajek J, Noszczyk-Nowak A. Biological Potential of Polyphenols in the Context of Metabolic Syndrome: An Analysis of Studies on Animal Models. BIOLOGY 2022; 11:biology11040559. [PMID: 35453758 PMCID: PMC9029039 DOI: 10.3390/biology11040559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023]
Abstract
Metabolic syndrome (MetS) is a disease that has a complex etiology. It is defined as the co-occurrence of several pathophysiological disorders, including obesity, hyperglycemia, hypertension, and dyslipidemia. MetS is currently a severe problem in the public health care system. As its prevalence increases every year, it is now considered a global problem among adults and young populations. The treatment of choice comprises lifestyle changes based mainly on diet and physical activity. Therefore, researchers have been attempting to discover new substances that could help reduce or even reverse the symptoms when added to food. These attempts have resulted in numerous studies. Many of them have investigated the bioactive potential of polyphenols as a "possible remedy", stemming from their antioxidative and anti-inflammatory effects and properties normalizing carbohydrate and lipid metabolism. Polyphenols may be supportive in preventing or delaying the onset of MetS or its complications. Additionally, the consumption of food rich in polyphenols should be considered as a supplement for antidiabetic drugs. To ensure the relevance of the studies on polyphenols' properties, mechanisms of action, and potential human health benefits, researchers have used laboratory animals displaying pathophysiological changes specific to MetS. Polyphenols or their plant extracts were chosen according to the most advantageous mitigation of pathological changes in animal models best reflecting the components of MetS. The present paper comprises an overview of animal models of MetS, and promising polyphenolic compounds whose bioactive potential, effect on metabolic pathways, and supplementation-related benefits were analyzed based on in vivo animal models.
Collapse
Affiliation(s)
- Joanna Niewiadomska
- Doctoral School of Wroclaw, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
- Correspondence:
| | | | - Jacek Gajek
- Department of Emergency Medical Service, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Agnieszka Noszczyk-Nowak
- Department of Internal and Diseases with Clinic for Horses, Dogs, and Cats, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland;
| |
Collapse
|
23
|
Doukbi E, Soghomonian A, Sengenès C, Ahmed S, Ancel P, Dutour A, Gaborit B. Browning Epicardial Adipose Tissue: Friend or Foe? Cells 2022; 11:991. [PMID: 35326442 PMCID: PMC8947372 DOI: 10.3390/cells11060991] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
Abstract
The epicardial adipose tissue (EAT) is the visceral fat depot of the heart which is highly plastic and in direct contact with myocardium and coronary arteries. Because of its singular proximity with the myocardium, the adipokines and pro-inflammatory molecules secreted by this tissue may directly affect the metabolism of the heart and coronary arteries. Its accumulation, measured by recent new non-invasive imaging modalities, has been prospectively associated with the onset and progression of coronary artery disease (CAD) and atrial fibrillation in humans. Recent studies have shown that EAT exhibits beige fat-like features, and express uncoupling protein 1 (UCP-1) at both mRNA and protein levels. However, this thermogenic potential could be lost with age, obesity and CAD. Here we provide an overview of the physiological and pathophysiological relevance of EAT and further discuss whether its thermogenic properties may serve as a target for obesity therapeutic management with a specific focus on the role of immune cells in this beiging phenomenon.
Collapse
Affiliation(s)
- Elisa Doukbi
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
| | - Astrid Soghomonian
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, F-13005 Marseille, France
| | - Coralie Sengenès
- Stromalab, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, University of Toulouse, F-31100 Toulouse, France;
- Institut National de la Santé et de la Recherche Médicale, University Paul Sabatier, F-31100 Toulouse, France
| | - Shaista Ahmed
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
| | - Patricia Ancel
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
| | - Anne Dutour
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, F-13005 Marseille, France
| | - Bénédicte Gaborit
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, F-13005 Marseille, France
| |
Collapse
|
24
|
Okamoto C, Tsukamoto O, Hasegawa T, Hitsumoto T, Matsuoka K, Takashima S, Amaki M, Kanzaki H, Izumi C, Ito S, Kitakaze M. Lower B-type natriuretic peptide levels predict left ventricular concentric remodelling and insulin resistance. ESC Heart Fail 2021; 9:636-647. [PMID: 34786876 PMCID: PMC8787986 DOI: 10.1002/ehf2.13700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/21/2021] [Accepted: 10/29/2021] [Indexed: 01/19/2023] Open
Abstract
Aims Natriuretic peptides have reportedly been associated with cardiac hypertrophy and insulin resistance; however, it has not been established if B‐type natriuretic peptide (BNP) is associated with either insulin resistance or cardiac remodelling in a population with normal plasma BNP levels. We investigated the relationship among plasma BNP levels, insulin resistance, and left ventricular (LV) remodelling in a population with normal physiological plasma BNP levels. Methods and results Among 1632 individuals who participated in annual health checks between 2005 and 2008 in Arita‐cho, Saga, Japan, 675 individuals [median (interquartile range) for age 62 (51–69) years; 227 men (34%)] with LV ejection fraction 50% and BNP level <35 pg/mL were enrolled in this study. Insulin resistance was assessed using homeostatic model assessment of insulin resistance (HOMA‐IR). LV geometry, including LV concentric remodelling, was classified based on relative wall thickness (RWT) and LV mass index values derived from echocardiographic findings. Factors associated with insulin resistance and LV geometry were investigated using multiple logistic regression analysis. Tertiles of BNP were inversely associated with HOMA‐IR [1st tertile, 1.33 (0.76–1.74); 2nd tertile, 1.05 (0.72–1.59); 3rd tertile, 0.95 (0.66–1.58), P = 0.005]. Lower BNP was associated with the prevalence of insulin resistance, defined as HOMA‐IR ≥1.37, even after full multivariate adjustment [1 SD increment in BNP = adjusted odds ratio (aOR) 0.740; 95% confidence interval (CI) 0.601–0.912; P = 0.005]. LV concentric remodelling (RWT >0.42; LV mass index ≤115 g/m2 in men and ≤95 g/m2 in women) was observed in 107 (16%) participants, while normal LV geometry (RWT ≤0.42; LV mass index ≤115 g/m2 in men and ≤95 g/m2 in women) was seen in 423 (63%), and LV hypertrophy (LV mass index >115 g/m2 in men and >95 g/m2 in women) in 145 (21%). Both low BNP level and higher insulin resistance were independently linked to LV concentric remodelling after multivariate adjustment (1 SD increment in BNP = aOR 0.714, 95% CI 0.544–0.938, P = 0.015; HOMA‐IR ≥ 1.37 vs. <1.37: aOR 1.694, 95% CI 1.004–2.857, P = 0.048, respectively). Conclusions Lower BNP levels are linked to either insulin resistance or LV concentric remodelling in a population with normal plasma BNP levels, suggesting that participants with lower natriuretic peptide level might be vulnerable to the development of metabolic disorders and LV morphological abnormalities.
Collapse
Affiliation(s)
- Chisato Okamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Osamu Tsukamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takuya Hasegawa
- Department of Cardiovascular Medicine, Garacia Hospital, Mino, Osaka, Japan
| | - Tatsuro Hitsumoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ken Matsuoka
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Makoto Amaki
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Hideaki Kanzaki
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Chisato Izumi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Shin Ito
- Department of Clinical Medicine and Development, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Masafumi Kitakaze
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Clinical Medicine and Development, National Cerebral and Cardiovascular Center, Osaka, Japan.,Hanwa Daini Senboku Hospital, Sakai, Osaka, 599-8271, Japan
| |
Collapse
|
25
|
Mishra S, Sadagopan N, Dunkerly-Eyring B, Rodriguez S, Sarver DC, Ceddia RP, Murphy SA, Knutsdottir H, Jani VP, Ashok D, Oeing CU, O'Rourke B, Gangoiti JA, Sears DD, Wong GW, Collins S, Kass DA. Inhibition of phosphodiesterase type 9 reduces obesity and cardiometabolic syndrome in mice. J Clin Invest 2021; 131:148798. [PMID: 34618683 DOI: 10.1172/jci148798] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 09/16/2021] [Indexed: 12/16/2022] Open
Abstract
Central obesity with cardiometabolic syndrome (CMS) is a major global contributor to human disease, and effective therapies are needed. Here, we show that cyclic GMP-selective phosphodiesterase 9A inhibition (PDE9-I) in both male and ovariectomized female mice suppresses preestablished severe diet-induced obesity/CMS with or without superimposed mild cardiac pressure load. PDE9-I reduces total body, inguinal, hepatic, and myocardial fat; stimulates mitochondrial activity in brown and white fat; and improves CMS, without significantly altering activity or food intake. PDE9 localized at mitochondria, and its inhibition in vitro stimulated lipolysis in a PPARα-dependent manner and increased mitochondrial respiration in both adipocytes and myocytes. PPARα upregulation was required to achieve the lipolytic, antiobesity, and metabolic effects of PDE9-I. All these PDE9-I-induced changes were not observed in obese/CMS nonovariectomized females, indicating a strong sexual dimorphism. We found that PPARα chromatin binding was reoriented away from fat metabolism-regulating genes when stimulated in the presence of coactivated estrogen receptor-α, and this may underlie the dimorphism. These findings have translational relevance given that PDE9-I is already being studied in humans for indications including heart failure, and efficacy against obesity/CMS would enhance its therapeutic utility.
Collapse
Affiliation(s)
| | | | | | - Susana Rodriguez
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ryan P Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Hildur Knutsdottir
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Vivek P Jani
- Division of Cardiology, Department of Medicine, and.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | - Jon A Gangoiti
- UCSD Biochemical Genetics and Metabolomics Laboratory and
| | - Dorothy D Sears
- Department of Medicine, UCSD, La Jolla, California, USA.,College of Health Solutions, Arizona State University, Phoenix, Arizona, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - David A Kass
- Division of Cardiology, Department of Medicine, and.,Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
26
|
Campolo F, Pofi R, Venneri MA, Isidori AM. Priming metabolism with the type 5 phosphodiesterase: the role of cGMP-hydrolyzing enzymes. Curr Opin Pharmacol 2021; 60:298-305. [PMID: 34507030 DOI: 10.1016/j.coph.2021.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/24/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022]
Abstract
The cyclic guanosine monophosphate (cGMP) signaling system is one of the most prominent regulators of many physiopathological processes in humans and rodents. It has been strongly established as an accomplished cellular signal involved in the regulation of energy homeostasis and cell metabolism, and pharmacological enhancement of cGMP has shown beneficial effects in metabolic disorders models. cGMP intracellular levels are finely regulated by phosphodiesterases (PDEs). The main enzyme responsible for the degradation of cGMP is PDE5. Preclinical and clinical studies have shown that PDE5 inhibitors (PDE5i) have beneficial effects on improving insulin resistance and glucose metabolism representing a promising therapeutic strategy for the treatment of metabolic disorders. This review aims to describe the molecular basis underlying the use of PDE5i to prompt cell metabolism and summarize current clinical trials assessing the effects of PDE5i on glucose metabolism.
Collapse
Affiliation(s)
- Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Riccardo Pofi
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Italy.
| |
Collapse
|
27
|
Treatment with atrial natriuretic peptide induces adipose tissue browning and exerts thermogenic actions in vivo. Sci Rep 2021; 11:17466. [PMID: 34465848 PMCID: PMC8408225 DOI: 10.1038/s41598-021-96970-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/18/2021] [Indexed: 01/14/2023] Open
Abstract
Increasing evidence suggests natriuretic peptides (NPs) coordinate inter-organ metabolic crosstalk with adipose tissues and play a critical role in energy metabolism. We recently reported A-type NP (ANP) raises intracellular temperature in cultured adipocytes in a low-temperature-sensitive manner. We herein investigated whether exogenous ANP-treatment exerts a significant impact on adipose tissues in vivo. Mice fed a high-fat-diet (HFD) or normal-fat-diet (NFD) for 13 weeks were treated with or without ANP infusion subcutaneously for another 3 weeks. ANP-treatment significantly ameliorated HFD-induced insulin resistance. HFD increased brown adipose tissue (BAT) cell size with the accumulation of lipid droplets (whitening), which was suppressed by ANP-treatment (re-browning). Furthermore, HFD induced enlarged lipid droplets in inguinal white adipose tissue (iWAT), crown-like structures in epididymal WAT, and hepatic steatosis, all of which were substantially attenuated by ANP-treatment. Likewise, ANP-treatment markedly increased UCP1 expression, a specific marker of BAT, in iWAT (browning). ANP also further increased UCP1 expression in BAT with NFD. Accordingly, cold tolerance test demonstrated ANP-treated mice were tolerant to cold exposure. In summary, exogenous ANP administration ameliorates HFD-induced insulin resistance by attenuating hepatic steatosis and by inducing adipose tissue browning (activation of the adipose tissue thermogenic program), leading to in vivo thermogenesis during cold exposure.
Collapse
|
28
|
da Silva MS, Lazo M, Daya NR, Tang O, Schaan BD, Ballantyne CM, Ndumele C, Selvin E. Six-year changes in N-terminal pro-brain natriuretic peptide and changes in weight and risk of obesity. Obesity (Silver Spring) 2021; 29:1215-1222. [PMID: 34159759 PMCID: PMC8231730 DOI: 10.1002/oby.23181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 11/05/2022]
Abstract
OBJECTIVE The aim of this study was to study the prospective association between N-terminal pro-brain natriuretic peptide (NT-proBNP) and changes in weight and obesity risk in a community-based population. METHODS Data from 9,681 participants from the Atherosclerosis Risk in Communities Study were analyzed at two time points 6 years apart. Among people without obesity at baseline, multivariable logistic regression models were used to examine the association between baseline levels of NT-proBNP and incident obesity. A multivariable linear regression model was used to examine the association between changes in NT-proBNP (visit 2 serum and visit 4 plasma samples) and changes in weight. RESULTS The prevalence of obesity increased from 28% to 35% in the 6-year follow-up period. Compared with individuals in the highest NT-proBNP quartile, those in the lowest were more likely to have obesity at baseline (odds ratio 1.25; 95% CI: 1.08-1.45) and, among people who did not have obesity at baseline, were more likely to develop obesity at follow-up (odds ratio 1.35; 95% CI: 1.07-1.69). Changes in NT-proBNP were inversely associated with weight change. CONCLUSIONS In this prospective study, lower levels of NT-proBNP were associated with higher risk of obesity, and changes in NT-proBNP were inversely associated with changes in weight. This suggests that natriuretic peptides or their pathways may be potential targets in the treatment of obesity.
Collapse
Affiliation(s)
- Mariana Sbaraini da Silva
- Postgraduate Program in Cardiology and Cardiovascular Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Mariana Lazo
- Department of Medicine, Division of General Internal Medicine, Johns Hopkins University, Baltimore, MD
- Urban Health Collaborative, Dornsife School of Public Health, Drexel University, Philadelphia, PA, USA
| | - Natalie R. Daya
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Olive Tang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Beatriz D. Schaan
- Postgraduate Program in Cardiology and Cardiovascular Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Hospital de Clinicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Christie M. Ballantyne
- Department of Medicine, Baylor College of Medicine, and Methodist DeBakey Heart and Vascular Center, Houston, TX, USA
| | - Chiadi Ndumele
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
29
|
Mishra S, Kass DA. Cellular and molecular pathobiology of heart failure with preserved ejection fraction. Nat Rev Cardiol 2021; 18:400-423. [PMID: 33432192 PMCID: PMC8574228 DOI: 10.1038/s41569-020-00480-6] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2020] [Indexed: 01/30/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) affects half of all patients with heart failure worldwide, is increasing in prevalence, confers substantial morbidity and mortality, and has very few effective treatments. HFpEF is arguably the greatest unmet medical need in cardiovascular disease. Although HFpEF was initially considered to be a haemodynamic disorder characterized by hypertension, cardiac hypertrophy and diastolic dysfunction, the pandemics of obesity and diabetes mellitus have modified the HFpEF syndrome, which is now recognized to be a multisystem disorder involving the heart, lungs, kidneys, skeletal muscle, adipose tissue, vascular system, and immune and inflammatory signalling. This multiorgan involvement makes HFpEF difficult to model in experimental animals because the condition is not simply cardiac hypertrophy and hypertension with abnormal myocardial relaxation. However, new animal models involving both haemodynamic and metabolic disease, and increasing efforts to examine human pathophysiology, are revealing new signalling pathways and potential therapeutic targets. In this Review, we discuss the cellular and molecular pathobiology of HFpEF, with the major focus being on mechanisms relevant to the heart, because most research has focused on this organ. We also highlight the involvement of other important organ systems, including the lungs, kidneys and skeletal muscle, efforts to characterize patients with the use of systemic biomarkers, and ongoing therapeutic efforts. Our objective is to provide a roadmap of the signalling pathways and mechanisms of HFpEF that are being characterized and which might lead to more patient-specific therapies and improved clinical outcomes.
Collapse
Affiliation(s)
- Sumita Mishra
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David A. Kass
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,
| |
Collapse
|
30
|
Hollstein T, Schlicht K, Krause L, Hagen S, Rohmann N, Schulte DM, Türk K, Beckmann A, Ahrens M, Franke A, Schreiber S, Becker T, Beckmann J, Laudes M. Effect of various weight loss interventions on serum NT-proBNP concentration in severe obese subjects without clinical manifest heart failure. Sci Rep 2021; 11:10096. [PMID: 33980890 PMCID: PMC8115663 DOI: 10.1038/s41598-021-89426-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 04/22/2021] [Indexed: 11/10/2022] Open
Abstract
Obesity is associated with a “natriuretic handicap” indicated by reduced N-terminal fragment of proBNP (NT-proBNP) concentration. While gastric bypass surgery improves the natriuretic handicap, it is presently unclear if sleeve gastrectomy exhibits similar effects. We examined NT-proBNP serum concentration in n = 72 obese participants without heart failure before and 6 months after sleeve gastrectomy (n = 28), gastric bypass surgery (n = 19), and 3-month 800 kcal/day very-low calorie diet (n = 25). A significant weight loss was observed in all intervention groups. Within 6 months, NT-proBNP concentration tended to increase by a median of 44.3 pg/mL in the sleeve gastrectomy group (p = 0.07), while it remained unchanged in the other groups (all p ≥ 0.50). To gain insights into potential effectors, we additionally analyzed NT-proBNP serum concentration in n = 387 individuals with different metabolic phenotypes. Here, higher NT-proBNP levels were associated with lower nutritional fat and protein but not with carbohydrate intake. Of interest, NT-proBNP serum concentrations were inversely correlated with fasting glucose concentration in euglycemic individuals but not in individuals with prediabetes or type 2 diabetes. In conclusion, sleeve gastrectomy tended to increase NT-proBNP levels in obese individuals and might improve the obesity-associated “natriuretic handicap”. Thereby, nutritional fat and protein intake and the individual glucose homeostasis might be metabolic determinants of NT-proBNP serum concentration.
Collapse
Affiliation(s)
- Tim Hollstein
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany
| | - Kristina Schlicht
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany
| | - Laura Krause
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany
| | - Stefanie Hagen
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany
| | - Nathalie Rohmann
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany
| | - Dominik M Schulte
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany
| | - Kathrin Türk
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany
| | - Alexia Beckmann
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany
| | - Markus Ahrens
- Helios Klinik Lengerich, Martin-Luther-Straße 49, 49525, Lengerich, Germany
| | - Andre Franke
- Institute for Clinical Molecular Biology, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany
| | - Stefan Schreiber
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany.,Institute for Clinical Molecular Biology, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany
| | - Thomas Becker
- Department of General and Abdominal Surgery, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany
| | - Jan Beckmann
- Department of General and Abdominal Surgery, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany
| | - Matthias Laudes
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Arnold Heller Straße 3, 24105, Kiel, Germany.
| |
Collapse
|
31
|
Song E, Da Eira D, Jani S, Sepa-Kishi D, Vu V, Hunter H, Lai M, Wheeler MB, Ceddia RB, Sweeney G. Cardiac Autophagy Deficiency Attenuates ANP Production and Disrupts Myocardial-Adipose Cross Talk, Leading to Increased Fat Accumulation and Metabolic Dysfunction. Diabetes 2021; 70:51-61. [PMID: 33046483 DOI: 10.2337/db19-0762] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/01/2020] [Indexed: 12/09/2022]
Abstract
Increased myocardial autophagy has been established as an important stress-induced cardioprotective response. Three weeks after generating cardiomyocyte-specific autophagy deficiency, via inducible deletion of autophagy-related protein 7 (Atg7), we found that these mice (AKO) had increased body weight and fat mass without altered food intake. Glucose and insulin tolerance tests indicated reduced insulin sensitivity in AKO mice. Metabolic cage analysis showed reduced ambulatory activity and oxygen consumption with a trend of elevated respiratory exchange ratio in AKO mice. Direct analysis of metabolism in subcutaneous and visceral adipocytes showed increased glucose oxidation and reduced ATGL expression and HSL phosphorylation with no change in lipid synthesis or fatty acid oxidation. Importantly, we found AKO mice had reduced myocardial and circulating levels of atrial natriuretic peptide (ANP), an established mediator of myocardial-adipose cross talk. When normal ANP levels were restored to AKO mice with use of osmotic pump, the metabolic dysfunction evident in AKO mice was corrected. We conclude that cardiac autophagy deficiency alters myocardial-adipose cross talk via decreased ANP levels with adverse metabolic consequences.
Collapse
Affiliation(s)
- Erfei Song
- Department of Biology, York University, Toronto, Canada
- The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Daniel Da Eira
- School of Kinesiology and Health Science, York University, Toronto, Canada
| | - Shailee Jani
- School of Kinesiology and Health Science, York University, Toronto, Canada
| | - Diane Sepa-Kishi
- School of Kinesiology and Health Science, York University, Toronto, Canada
| | - Vivian Vu
- Department of Biology, York University, Toronto, Canada
| | - Howard Hunter
- Department of Chemistry, York University, Toronto, Canada
| | - Mi Lai
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Michael B Wheeler
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada
- University Health Network, Toronto, Canada
| | - Rolando B Ceddia
- School of Kinesiology and Health Science, York University, Toronto, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Canada
| |
Collapse
|
32
|
Hahn VS, Knutsdottir H, Luo X, Bedi K, Margulies KB, Haldar SM, Stolina M, Yin J, Khakoo AY, Vaishnav J, Bader JS, Kass DA, Sharma K. Myocardial Gene Expression Signatures in Human Heart Failure With Preserved Ejection Fraction. Circulation 2020; 143:120-134. [PMID: 33118835 DOI: 10.1161/circulationaha.120.050498] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Heart failure (HF) with preserved ejection fraction (HFpEF) constitutes half of all HF but lacks effective therapy. Understanding of its myocardial biology remains limited because of a paucity of heart tissue molecular analysis. METHODS We performed RNA sequencing on right ventricular septal endomyocardial biopsies prospectively obtained from patients meeting consensus criteria for HFpEF (n=41) contrasted with right ventricular septal tissue from patients with HF with reduced ejection fraction (HFrEF, n=30) and donor controls (n=24). Principal component analysis and hierarchical clustering tested for transcriptomic distinctiveness between groups, effect of comorbidities, and differential gene expression with pathway enrichment contrasted HF groups and donor controls. Within HFpEF, non-negative matrix factorization and weighted gene coexpression analysis identified molecular subgroups, and the resulting clusters were correlated with hemodynamic and clinical data. RESULTS Patients with HFpEF were more often women (59%), African American (68%), obese (median body mass index 41), and hypertensive (98%), with clinical HF characterized by 65% New York Heart Association Class III or IV, nearly all on a loop diuretic, and 70% with a HF hospitalization in the previous year. Principal component analysis separated HFpEF from HFrEF and donor controls with minimal overlap, and this persisted after adjusting for primary comorbidities: body mass index, sex, age, diabetes, and renal function. Hierarchical clustering confirmed group separation. Nearly half the significantly altered genes in HFpEF versus donor controls (1882 up, 2593 down) changed in the same direction in HFrEF; however, 5745 genes were uniquely altered between HF groups. Compared with controls, uniquely upregulated genes in HFpEF were enriched in mitochondrial adenosine triphosphate synthesis/electron transport, pathways downregulated in HFrEF. HFpEF-specific downregulated genes engaged endoplasmic reticulum stress, autophagy, and angiogenesis. Body mass index differences largely accounted for HFpEF upregulated genes, whereas neither this nor broader comorbidity adjustment altered pathways enriched in downregulated genes. Non-negative matrix factorization identified 3 HFpEF transcriptomic subgroups with distinctive pathways and clinical correlates, including a group closest to HFrEF with higher mortality, and a mostly female group with smaller hearts and proinflammatory signaling. These groupings remained after sex adjustment. Weighted gene coexpression analysis yielded analogous gene clusters and clinical groupings. CONCLUSIONS HFpEF exhibits distinctive broad transcriptomic signatures and molecular subgroupings with particular clinical features and outcomes. The data reveal new signaling targets to consider for precision therapeutics.
Collapse
Affiliation(s)
- Virginia S Hahn
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (V.S.H., J.V., D.A.K., K.S.)
| | - Hildur Knutsdottir
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD (H.K., J.S.B., D.A.K.)
| | - Xin Luo
- Genome Analysis Unit, Amgen Research, San Francisco, CA (X.L., J.Y.)
| | - Kenneth Bedi
- Division of Cardiology, University of Pennsylvania, Philadelphia (K.B., K.B.M.)
| | - Kenneth B Margulies
- Division of Cardiology, University of Pennsylvania, Philadelphia (K.B., K.B.M.)
| | - Saptarsi M Haldar
- Cardiometabolic Disorders Research, Amgen Research, San Francisco, CA (S.M.H., A.Y.K.)
| | - Marina Stolina
- Cardiometabolic Disorders Research, Amgen Research, Thousand Oaks, CA (M.S.)
| | - Jun Yin
- Genome Analysis Unit, Amgen Research, San Francisco, CA (X.L., J.Y.)
| | - Aarif Y Khakoo
- Cardiometabolic Disorders Research, Amgen Research, San Francisco, CA (S.M.H., A.Y.K.).,Calico Life Sciences, San Francisco, CA (A.Y.K.)
| | - Joban Vaishnav
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (V.S.H., J.V., D.A.K., K.S.)
| | - Joel S Bader
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD (H.K., J.S.B., D.A.K.)
| | - David A Kass
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (V.S.H., J.V., D.A.K., K.S.).,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD (H.K., J.S.B., D.A.K.)
| | - Kavita Sharma
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (V.S.H., J.V., D.A.K., K.S.)
| |
Collapse
|
33
|
Wu G, Liu Y, Feng W, An X, Lin W, Tang C. Hypoxia-Induced Adipose Lipolysis Requires Fibroblast Growth Factor 21. Front Pharmacol 2020; 11:1279. [PMID: 32922298 PMCID: PMC7456904 DOI: 10.3389/fphar.2020.01279] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/03/2020] [Indexed: 02/05/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a recently discovered hepatokine that regulates lipid and glucose metabolism and is upregulated in response to numerous physiological and pathological stimuli. Herein, we demonstrate that both physical and chemical hypoxia increase the systemic and hepatic expression of FGF21 in mice; by contrast, hypoxia induces a reduction of FGF21 expression in hepatocytes, indicating that hypoxia-induced FGF21 expression is differentially regulated in intact animals and in hepatocytes. Furthermore, we demonstrate that hypoxia treatment increases hormone-sensitive lipase-mediated adipose tissue lipolysis in mice, which is reduced in Fgf21 knockout mice, thereby implying that FGF21 plays a critical role in hypoxia-related adipose lipolysis. Adipose tissue lipolysis causes an increase in the amount of circulating free fatty acids, which leads to the activation of peroxisome proliferators-activated receptor alpha and an increased expression of FGF21 in hepatocytes. We further show that hypoxia-induced elevation of reactive oxygen species, but not the hypoxia-inducible factor, is responsible for the lipolysis and FGF21 expression. In conclusion, our data clearly demonstrate that FGF21 plays a critical role in hypoxia-induced adipose lipolysis, which induces hepatic expression of FGF21. Clarification of hypoxia-regulated FGF21 regulation will enhance our understanding of the pathophysiology of hypoxia-related diseases, such as sleep disorders and metabolic diseases.
Collapse
Affiliation(s)
- Guicheng Wu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China.,Department of Hepatology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Yanlong Liu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.,Zhuji Institute of Biomedicine, School of Pharmaceutical Sciences, Wenzhou Medical University, Shaoxing, China
| | - Wenke Feng
- Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, United States
| | - Xuan An
- Department of Hepatology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Wenhui Lin
- Department of Cardiology, Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, China
| | - Chengwei Tang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, Chengdu, China
| |
Collapse
|
34
|
Kang R, Nagoshi T, Kimura H, Tanaka TD, Yoshii A, Inoue Y, Morimoto S, Ogawa K, Minai K, Ogawa T, Kawai M, Yoshimura M. Possible Association Between Body Temperature and B-Type Natriuretic Peptide in Patients With Cardiovascular Diseases. J Card Fail 2020; 27:75-82. [PMID: 32871239 DOI: 10.1016/j.cardfail.2020.08.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 07/31/2020] [Accepted: 08/24/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND In addition to various biological effects of natriuretic peptides (NP) on cardiovascular systems, we recently reported that NP raises intracellular temperature in cultured adipocytes. We herein examined the possible thermogenic action of NP in consideration of hemodynamic parameters and inflammatory reaction by proposing structural equation models. METHODS AND RESULTS The study population consisted of 1985 consecutive patients who underwent cardiac catheterization. Covariance structure analyses were performed to clarify the direct contribution of plasma B-type NP (BNP) to body temperature (BT) by excluding other confounding factors. A hierarchical path model showed increase in BNP, increase in C-reactive protein and decrease in left ventricular ejection fraction were mutually associated. As expected, C-reactive protein was positively correlated with BT. Importantly, despite a negative correlation between BNP and left ventricular ejection fraction, a decrease in the left ventricular ejection fraction was associated with BT decrease, whereas elevation in BNP level was associated with BT increase independently of C-reactive protein level (P = .007). CONCLUSIONS Patients with LV dysfunction tend to manifest a decrease in BT, whereas BNP elevation is associated with an increase in BT independently of inflammatory response. These findings suggest the adaptive heat-retaining property of NP (and/or NP-associated factors) when BT falls owing to unfavorable hemodynamic conditions in a state of impaired cardiac function.
Collapse
Affiliation(s)
- Ryeonshi Kang
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Tomohisa Nagoshi
- 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
| | - Toshikazu D Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Akira Yoshii
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Yasunori Inoue
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Satoshi Morimoto
- 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
|
35
|
Icard P, Ollivier L, Forgez P, Otz J, Alifano M, Fournel L, Loi M, Thariat J. Perspective: Do Fasting, Caloric Restriction, and Diets Increase Sensitivity to Radiotherapy? A Literature Review. Adv Nutr 2020; 11:1089-1101. [PMID: 32492154 PMCID: PMC7490158 DOI: 10.1093/advances/nmaa062] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/11/2020] [Accepted: 05/06/2020] [Indexed: 12/17/2022] Open
Abstract
Caloric starvation, as well as various diets, has been proposed to increase the oxidative DNA damage induced by radiotherapy (RT). However, some diets could have dual effects, sometimes promoting cancer growth, whereas proposing caloric restriction may appear counterproductive during RT considering that the maintenance of weight is a major factor for the success of this therapy. A systematic review was performed via a PubMed search on RT and fasting, or caloric restriction, ketogenic diet (>75% of fat-derived energy intake), protein starvation, amino acid restriction, as well as the Warburg effect. Twenty-six eligible original articles (17 preclinical studies and 9 clinical noncontrolled studies on low-carbohydrate, high-fat diets popularized as ketogenic diets, representing a total of 77 patients) were included. Preclinical experiments suggest that a short period of fasting prior to radiation, and/or transient caloric restriction during treatment course, can increase tumor responsiveness. These regimens promote accumulation of oxidative lesions and insufficient repair, subsequently leading to cancer cell death. Due to their more flexible metabolism, healthy cells should be less sensitive, shifting their metabolism to support survival and repair. Interestingly, these regimens might stimulate an acute anticancer immune response, and may be of particular interest in tumors with high glucose uptake on positron emission tomography scan, a phenotype associated with poor survival and resistance to RT. Preclinical studies with ketogenic diets yielded more conflicting results, perhaps because cancer cells can sometimes metabolize fatty acids and/or ketone bodies. Randomized trials are awaited to specify the role of each strategy according to the clinical setting, although more stringent definitions of proposed diet, nutritional status, and consensual criteria for tumor response assessment are needed. In conclusion, dietary interventions during RT could be a simple and medically economical and inexpensive method that may deserve to be tested to improve efficiency of radiation.
Collapse
Affiliation(s)
- Philippe Icard
- Université Caen Normandie, Normandie University, UNICAEN, Medical School, CHU de Caen, Caen, France,Inserm U1086 Interdisciplinary Research Unit for Cancer Prevention and Treatment, Centre de Lutte Contre le Cancer, Centre François Baclesse, Caen, France,Service de Chirurgie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, APHP, Paris-Descartes University, Paris, France,Address correspondence to PI (e-mail: )
| | - Luc Ollivier
- Centre Hospitalier de Brest, Université de Bretagne Occidentale, Brest, France,Centre François Baclesse, Radiotherapy Unit, Caen, France
| | - Patricia Forgez
- INSERM UMR-S 1124, Cellular Homeostasis and Cancer, Paris-Descartes University, Paris, France
| | - Joelle Otz
- Department of Radiation Oncology, Institut Curie, Paris, France
| | - Marco Alifano
- Service de Chirurgie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, APHP, Paris-Descartes University, Paris, France,INSERM U1138, Integrative Cancer Immunology, University Paris Descartes, Paris, France
| | - Ludovic Fournel
- Service de Chirurgie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, APHP, Paris-Descartes University, Paris, France,INSERM U1138, Integrative Cancer Immunology, University Paris Descartes, Paris, France
| | - Mauro Loi
- Department of Radiation Oncology, Paris Est University Hospitals, AP-HP, Paris, France
| | - Juliette Thariat
- Université Caen Normandie, Normandie University, UNICAEN, Medical School, CHU de Caen, Caen, France,Centre François Baclesse, Radiotherapy Unit, Caen, France,Laboratoire de Physique Corpusculaire, IN2P3, Normandie University/UNICAEN/CNRS, Caen, France
| |
Collapse
|
36
|
Abstract
Investigations into the mixed muscle-secretory phenotype of cardiomyocytes from the atrial appendages of the heart led to the discovery that these cells produce, in a regulated manner, two polypeptide hormones - the natriuretic peptides - referred to as atrial natriuretic factor or atrial natriuretic peptide (ANP) and brain or B-type natriuretic peptide (BNP), thereby demonstrating an endocrine function for the heart. Studies on the gene encoding ANP (NPPA) initiated the field of modern research into gene regulation in the cardiovascular system. Additionally, ANP and BNP were found to be the natural ligands for cell membrane-bound guanylyl cyclase receptors that mediate the effects of natriuretic peptides through the generation of intracellular cGMP, which interacts with specific enzymes and ion channels. Natriuretic peptides have many physiological actions and participate in numerous pathophysiological processes. Important clinical entities associated with natriuretic peptide research include heart failure, obesity and systemic hypertension. Plasma levels of natriuretic peptides have proven to be powerful diagnostic and prognostic biomarkers of heart disease. Development of pharmacological agents that are based on natriuretic peptides is an area of active research, with vast potential benefits for the treatment of cardiovascular disease.
Collapse
|
37
|
N-terminal Pro-B-Type Natriuretic Peptide and Malnutrition in Patients on Hemodialysis. Int J Nephrol 2020; 2020:9528014. [PMID: 32206350 PMCID: PMC7077038 DOI: 10.1155/2020/9528014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 02/11/2020] [Indexed: 11/17/2022] Open
Abstract
Natriuretic peptides, brain natriuretic peptide (BNP), and N-terminal probrain natriuretic peptide (NT-proBNP) are mainly known as diagnostic markers for heart failure with high diagnostic and prognostic values in the general population. In patients who are undergoing hemodialysis (HD), changes in NT-proBNP can be related to noncardiac problems such as fluid overload, inflammation, or malnutrition and can also be influenced by the dialysis characteristics. The current review aimed to summarize findings from studies on the association between NT-proBNP and malnutrition in HD patients. Articles published after 2009 and over a ten-year period were considered for inclusion. We first briefly discuss the traditional functions of NT-proBNP, and after, we describe the functions of this prohormone by focusing on its relation with protein energy wasting (PEW) in HD patients. Mechanisms that could explain these relationships were also discussed. Overall, 7 studies in which the investigation of the relations between NT-proBNP and nutritional status in HD patients were among the main objects were taken into account. NT-proBNP levels correlated with several factors described in the 4 categories of markers indicative of PEW (body mass and composition, muscle mass, biochemical criteria, and dietary intakes) and/or were associated with PEW. Interactions between several parameters could be involved in the association between NT-proBNP and malnutrition with a strong role of weight status. NT-proBNP is elevated in HD patients and is associated with malnutrition. Nevertheless, the prognostic value of NT-proBNP on nutritional status should be evaluated.
Collapse
|
38
|
Ceddia RP, Collins S. A compendium of G-protein-coupled receptors and cyclic nucleotide regulation of adipose tissue metabolism and energy expenditure. Clin Sci (Lond) 2020; 134:473-512. [PMID: 32149342 PMCID: PMC9137350 DOI: 10.1042/cs20190579] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/17/2020] [Accepted: 02/24/2020] [Indexed: 12/15/2022]
Abstract
With the ever-increasing burden of obesity and Type 2 diabetes, it is generally acknowledged that there remains a need for developing new therapeutics. One potential mechanism to combat obesity is to raise energy expenditure via increasing the amount of uncoupled respiration from the mitochondria-rich brown and beige adipocytes. With the recent appreciation of thermogenic adipocytes in humans, much effort is being made to elucidate the signaling pathways that regulate the browning of adipose tissue. In this review, we focus on the ligand-receptor signaling pathways that influence the cyclic nucleotides, cAMP and cGMP, in adipocytes. We chose to focus on G-protein-coupled receptor (GPCR), guanylyl cyclase and phosphodiesterase regulation of adipocytes because they are the targets of a large proportion of all currently available therapeutics. Furthermore, there is a large overlap in their signaling pathways, as signaling events that raise cAMP or cGMP generally increase adipocyte lipolysis and cause changes that are commonly referred to as browning: increasing mitochondrial biogenesis, uncoupling protein 1 (UCP1) expression and respiration.
Collapse
Affiliation(s)
- Ryan P Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| |
Collapse
|
39
|
Ferrero KM, Koch WJ. Metabolic Crosstalk between the Heart and Fat. Korean Circ J 2020; 50:379-394. [PMID: 32096362 PMCID: PMC7098822 DOI: 10.4070/kcj.2019.0400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 12/27/2019] [Indexed: 12/14/2022] Open
Abstract
It is now recognized that the heart can behave as a true endocrine organ, which can modulate the function of other tissues. Emerging evidence has shown that visceral fat is one such distant organ the heart communicates with. In fact, it appears that bi-directional crosstalk between adipose tissue and the myocardium is crucial to maintenance of normal function in both organs. In particular, factors secreted from the heart are now known to influence the metabolic activity of adipose tissue and other organs, as well as modulate the release of metabolic substrates and signaling molecules from the periphery. This review summarizes current knowledge regarding primary cardiokines and adipokines involved in heart-fat crosstalk, as well as implications of their dysregulation for cardiovascular health.
Collapse
Affiliation(s)
- Kimberly M Ferrero
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Walter J Koch
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Bhargava P, Janda J, Schnellmann RG. Elucidation of cGMP-dependent induction of mitochondrial biogenesis through PKG and p38 MAPK in the kidney. Am J Physiol Renal Physiol 2019; 318:F322-F328. [PMID: 31841384 DOI: 10.1152/ajprenal.00533.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Previous studies have shown that cGMP increases mitochondrial biogenesis (MB). Our laboratory has determined that formoterol and LY344864, agonists of the β2-adrenergic receptor and 5-HT1F receptor, respectively, signal MB in a soluble guanylyl cyclase (sGC)-dependent manner. However, the pathway between cGMP and MB produced by these pharmacological agents in renal proximal tubule cells (RPTCs) and the kidney has not been determined. In the present study, we showed that treatment of RPTCs with formoterol, LY344864, or riociguat, a sGC stimulator, induces MB through protein kinase G (PKG), a target of cGMP, and p38, an associated downstream target of PKG and a regulator of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) expression in RPTCs. We also examined if p38 plays a role in PGC-1α phosphorylation in vivo. Administration of l-skepinone, a potent and specific inhibitor of p38α and p38β, to naïve mice inhibited phosphorylated PGC-1α localization in the nuclear fraction of the renal cortex. Taken together, we demonstrated a pathway, sGC/cGMP/PKG/p38/PGC-1α, for pharmacological induction of MB and the importance of p38 in this pathway.
Collapse
Affiliation(s)
- Pallavi Bhargava
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Jaroslav Janda
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona.,Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona
| |
Collapse
|
41
|
Spannella F, Giulietti F, Bordicchia M, Burnett JC, Sarzani R. Association Between Cardiac Natriuretic Peptides and Lipid Profile: a Systematic Review and Meta-Analysis. Sci Rep 2019; 9:19178. [PMID: 31844088 PMCID: PMC6915780 DOI: 10.1038/s41598-019-55680-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/20/2019] [Indexed: 02/08/2023] Open
Abstract
Cardiac natriuretic peptides (NPs) play a fundamental role in maintaining cardiovascular (CV) and renal homeostasis. Moreover, they also affect glucose and lipid metabolism. We performed a systematic review and meta-analysis of studies investigating the association of NPs with serum lipid profile. A PubMed and Scopus search (2005–2018) revealed 48 studies reporting the association between NPs and components of lipid profile [total cholesterol (TC), low-density lipoprotein cholesterol (LDLc), high-density lipoprotein cholesterol (HDLc) and triglycerides (TG)]. Despite high inconsistency across studies, NPs levels were inversely associated with TC [k = 32; pooled r = −0.09; I2 = 90.26%], LDLc [k = 31; pooled r = −0.09; I2 = 82.38%] and TG [k = 46; pooled r = −0.11; I2 = 94.14%], while they were directly associated with HDLc [k = 41; pooled r = 0.06; I2 = 87.94%]. The relationship with LDLc, HDLc and TG lost significance if only studies on special populations (works including subjects with relevant acute or chronic conditions that could have significantly affected the circulating levels of NPs or lipid profile) or low-quality studies were taken into account. The present study highlights an association between higher NP levels and a favorable lipid profile. This confirms and extends our understanding of the metabolic properties of cardiac NPs and their potential in CV prevention.
Collapse
Affiliation(s)
- Francesco Spannella
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, Ancona, Italy.,Department of Clinical and Molecular Sciences, University "Politecnica delle Marche", Via Tronto 10/a, Ancona, Italy
| | - Federico Giulietti
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, Ancona, Italy.,Department of Clinical and Molecular Sciences, University "Politecnica delle Marche", Via Tronto 10/a, Ancona, Italy
| | - Marica Bordicchia
- Department of Clinical and Molecular Sciences, University "Politecnica delle Marche", Via Tronto 10/a, Ancona, Italy
| | - John C Burnett
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Mayo Clinic and Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Riccardo Sarzani
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, Ancona, Italy. .,Department of Clinical and Molecular Sciences, University "Politecnica delle Marche", Via Tronto 10/a, Ancona, Italy.
| |
Collapse
|
42
|
Klepac K, Yang J, Hildebrand S, Pfeifer A. RGS2: A multifunctional signaling hub that balances brown adipose tissue function and differentiation. Mol Metab 2019; 30:173-183. [PMID: 31767169 PMCID: PMC6807268 DOI: 10.1016/j.molmet.2019.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/12/2019] [Accepted: 09/28/2019] [Indexed: 12/28/2022] Open
Abstract
Objective Recruitment of brown adipose tissue (BAT) is a potential new strategy for increasing energy expenditure (EE) to treat obesity. G protein–coupled receptors (GPCRs) represent promising targets to activate BAT, as they are the major regulators of BAT biological function. To identify new regulators of GPCR signaling in BAT, we studied the role of Regulator of G protein Signaling 2 (RGS2) in brown adipocytes and BAT. Methods We combined pharmacological and genetic tools to investigate the role of RGS2 in BAT in vitro and in vivo. Adipocyte progenitors were isolated from wild-type (WT) and RGS2 knockout (RGS2−/−) BAT and differentiated to brown adipocytes. This approach was complemented with knockdown of RGS2 using lentiviral shRNAs (shRGS2). Adipogenesis was analyzed by Oil Red O staining and by determining the expression of adipogenic and thermogenic markers. Pharmacological modulators and fluorescence staining of F-acting stress fibers were employed to identify the underlying signaling pathways. In vivo, the activity of BAT was assessed by ex vivo lipolysis and by measuring whole-body EE by indirect calorimetry in metabolic cages. Results RGS2 is highly expressed in BAT, and treatment with cGMP—an important enhancer of brown adipocyte differentiation—further increased RGS2 expression. Loss of RGS2 strongly suppressed adipogenesis and the expression of thermogenic genes in brown adipocytes. Mechanistically, we found increased Gq/Rho/Rho kinase (ROCK) signaling in the absence of RGS2. Surprisingly, in vivo analysis revealed elevated BAT activity in RGS2-deficient mice that was caused by enhanced Gs/cAMP signaling. Conclusion Overall, RGS2 regulates two major signaling pathways in BAT: Gq and Gs. On the one hand, RGS2 promotes brown adipogenesis by counteracting the inhibitory action of Gq/Rho/ROCK signaling. On the other hand, RGS2 decreases the activity of BAT through the inhibition of Gs signaling and cAMP production. Thus, RGS2 might represent a stress modulator that protects BAT from overstimulation. RGS2 regulates brown adipose tissue (BAT) by inhibiting two major G protein-coupled receptor (GPCR) pathways – Gq and Gs. Deletion of RGS2 impairs the differentiation of murine brown adipocytes due to elevated Gq/Rho/ROCK signaling. In vivo, RGS2 knock-out mice show an increase in BAT lipolysis and whole-body energy expenditure.
Collapse
Affiliation(s)
- Katarina Klepac
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany; Research Training Group 1873, University of Bonn, 53127 Bonn, Germany.
| | - JuHee Yang
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany; Research Training Group 1873, University of Bonn, 53127 Bonn, Germany
| | - Staffan Hildebrand
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany; Research Training Group 1873, University of Bonn, 53127 Bonn, Germany; PharmaCenter, University of Bonn, 53127 Bonn, Germany.
| |
Collapse
|
43
|
Oh A, Okazaki R, Sam F, Valero-Muñoz M. Heart Failure With Preserved Ejection Fraction and Adipose Tissue: A Story of Two Tales. Front Cardiovasc Med 2019; 6:110. [PMID: 31428620 PMCID: PMC6687767 DOI: 10.3389/fcvm.2019.00110] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by signs and symptoms of heart failure in the presence of a normal left ventricular ejection fraction. Although it accounts for up to 50% of all clinical presentations of heart failure, there are no evidence-based therapies for HFpEF to reduce morbidity and mortality. Additionally there is a lack of mechanistic understanding about the pathogenesis of HFpEF. HFpEF is associated with many comorbidities (such as obesity, hypertension, type 2 diabetes, atrial fibrillation, etc.) and is coupled with both cardiac and extra-cardiac abnormalities. Large outcome trials and registries reveal that being obese is a major risk factor for HFpEF. There is increasing focus on investigating the link between obesity and HFpEF, and the role that the adipose tissue and the heart, and the circulating milieu play in development and pathogenesis of HFpEF. This review discusses features of the obese-HFpEF phenotype and highlights proposed mechanisms implicated in the inter-tissue communication between adipose tissue and the heart in obesity-associated HFpEF.
Collapse
Affiliation(s)
- Albin Oh
- Evans Department of Medicine, Boston Medical Center, Boston, MA, United States
| | - Ross Okazaki
- Boston University School of Medicine, Boston, MA, United States
| | - Flora Sam
- Evans Department of Medicine, Boston Medical Center, Boston, MA, United States
- Boston University School of Medicine, Boston, MA, United States
- Section of Cardiovascular Medicine, Boston Medical Center, Boston, MA, United States
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Maria Valero-Muñoz
- Boston University School of Medicine, Boston, MA, United States
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
44
|
Role of epicardial adipose tissue NPR-C in acute coronary syndrome. Atherosclerosis 2019; 286:79-87. [DOI: 10.1016/j.atherosclerosis.2019.05.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/28/2019] [Accepted: 05/08/2019] [Indexed: 12/12/2022]
|
45
|
Collaborative Activities of Noradrenaline and Natriuretic Peptide for Glucose Utilization in Patients with Acute Coronary Syndrome. Sci Rep 2019; 9:7822. [PMID: 31127136 PMCID: PMC6534620 DOI: 10.1038/s41598-019-44216-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 05/13/2019] [Indexed: 12/14/2022] Open
Abstract
Glucose is an important preferential substrate for energy metabolism during acute coronary syndrome (ACS) attack, although insulin resistance (IR) increases during ACS. Increasing evidence indicates that natriuretic peptides (NP) regulate glucose homeostasis. We investigated possible compensatory actions of NP in collaboration with other neurohumoral factors that facilitate glucose utilization during ACS. The study population consisted of 1072 consecutive cases with ischemic heart disease who underwent cardiac catheterization (ACS, n = 216; non-ACS, n = 856). Among ACS subjects, biochemical data after acute-phase treatment were available in 91 cases, defined as ACS-remission phase (ACS-rem). Path models based on covariance structure analyses were proposed to clarify the direct contribution of B-type NP (BNP) and noradrenaline to glucose and HOMA-IR levels while eliminating confounding biases. In non-ACS and ACS-rem subjects, although noradrenaline slightly increased glucose and/or HOMA-IR levels (P < 0.03), BNP did not significantly affect them. In contrast, in ACS subjects, high noradrenaline was a significant cause of increases in glucose and HOMA-IR levels (P < 0.001), whereas high BNP was a significant cause of decreases in both parameters (P < 0.005). These findings indicate that BNP and noradrenaline coordinately activate glucose metabolism during ACS, with noradrenaline increasing glucose levels, as an energy substrate, while BNP improves IR and promotes glucose utilization.
Collapse
|
46
|
Bai F, Tu T, Qin F, Ma Y, Liu N, Liu Y, Liao X, Zhou S, Liu Q. Quantitative proteomics of changes in succinylated proteins expression profiling in left appendages tissue from valvular heart disease patients with atrial fibrillation. Clin Chim Acta 2019; 495:345-354. [PMID: 31059701 DOI: 10.1016/j.cca.2019.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/15/2019] [Accepted: 05/02/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Previous studies have suggested that proteomic modifications are closely associated with cardiovascular diseases. The aim of this study was to identify potential mechanisms by profiling the changes in succinylated protein expression in left appendage tissues from patients with valvular heart disease and atrial fibrillation (AF). METHODS Using dimethyl labeling for relative and absolute quantification-coupled high-performance liquid chromatography-tandem mass spectrometry, we analyzed the proteomics profiles and succinylation events in 18 left atrial appendage tissue samples from patients who underwent cardiac valvular surgery, including nine patients with permanent AF and nine patients with sinus rhythm (SR). RESULTS In total, after setting the quantification ratio > 1.3 and < 1:1.3 representing the up- and downregulated cutoff values, respectively, 132 proteins were classified as targets of upregulation and 117 proteins as targets of downregulation. Within these proteins, 246 sites exhibited upregulated succinylation and 45 sites exhibited downregulated succinylation. Protein-protein interaction networks showed that the proteins exhibiting lysine succinylation and AF status were highly enriched in energy metabolism, extracellular matrix-related, and cellular structure-related proteins. These results were confirmed by western blot. CONCLUSIONS The differences in succinylation level of energy metabolism-related proteins indicates the possible involvement of these proteins in AF of valvular heart disease patients, and provide insight for further analysis of their biological functions.
Collapse
Affiliation(s)
- Fan Bai
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Tao Tu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Fen Qin
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yingxu Ma
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Na Liu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yaozhong Liu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiaobo Liao
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Shenghua Zhou
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qiming Liu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
47
|
Öztop M, Özbek M, Liman N, Beyaz F, Ergün E, Ergün L. Localization profiles of natriuretic peptides in hearts of pre-hibernating and hibernating Anatolian ground squirrels (Spermophilus xanthoprymnus). Vet Res Commun 2019; 43:45-65. [PMID: 30689110 DOI: 10.1007/s11259-019-9745-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 01/22/2019] [Indexed: 01/13/2023]
Abstract
The Anatolian ground squirrel (Spermophilus xanthoprymnus) is a typical example of true mammalian hibernators. In order to adapt to extreme external and internal environments during hibernation, they lower their body temperatures, heart rates and oxygen consumption; however, pathological events such as ischemia and ventricular fibrillation do not occur in their cardiovascular systems. During the hibernation, maintenance of cardiac function is very important for survival of ground squirrels. Natriuretic peptides (NPs) are key factors in the regulation of cardiovascular hemostasis. Since NPs' role on the protection of heart during hibernation are less clear, the aim of this study was to investigate dynamic changes in NPs content in the cardiac chambers and to reveal the possible role of NPs on establishing cardiac function in ground squirrel during hibernation using immunohistochemistry. The immunohistochemical results indicate that cardiac NP expressions in atrial and ventricular cardiomyocytes were different from each other and were sex-independent. ANP and BNP were expressed in a chamber-dependent manner in female and male squirrel hearts. Furthermore, cardiac NPs expression levels in hibernation period were lower than those at the pre-hibernation period. During prehibernation period, ANP, BNP and CNP were expressed in the white and beige adipocytes of epicardial adipose tissue (EAT); while during hibernation period, the brown adipocytes of EAT were positive for BNP and CNP. These data suggest that the hibernation-dependent reduction in levels of NPs, particularly ANP, in cardiac chambers and EAT may be associated with low heart rate and oxygen consumption during hibernation. However, further studies are needed to better delineate the roles of NPs during the hibernation.
Collapse
Affiliation(s)
- Mustafa Öztop
- Department of Biology, Faculty of Science and Art, Mehmet Akif Ersoy University, Burdur, Turkey.
| | - Mehmet Özbek
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Mehmet Akif Ersoy University, Burdur, Turkey
| | - Narin Liman
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Feyzullah Beyaz
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Emel Ergün
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| | - Levent Ergün
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
48
|
Prasad GVR, Yan AT, Nash MM, Kim SJ, Wald R, Wald R, Lok C, Gunaratnam L, Karur GR, Kirpalani A, Connelly PW. Determinants of Left Ventricular Characteristics Assessed by Cardiac Magnetic Resonance Imaging and Cardiovascular Biomarkers Related to Kidney Transplantation. Can J Kidney Health Dis 2018; 5:2054358118809974. [PMID: 30542623 PMCID: PMC6236646 DOI: 10.1177/2054358118809974] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/10/2018] [Indexed: 01/03/2023] Open
Abstract
Background: Cardiac magnetic resonance (CMR) imaging accurately and precisely measures
left ventricular (LV) mass and function. Identifying mechanisms by which LV
mass change and functional improvement occur in some end-stage kidney
disease (ESKD) patients may help to appropriately target kidney transplant
(KT) recipients for further investigation and intervention. The
concentration of serum adiponectin, a cardiovascular biomarker, increases in
cardiac failure, its production being enhanced by B-type natriuretic peptide
(BNP), and both serum adiponectin and BNP concentrations decline
posttransplantation. Objective: We tested the hypothesis that kidney transplantation alters LV
characteristics that relate to serum adiponectin concentrations. Design: Prospective and observational cohort study. Setting: The study was performed at 3 adult kidney transplant and dialysis centers in
Ontario, Canada. Patients: A total of 82 KT candidate subjects were recruited (39 to the KT group and 43
to the dialysis group). Predialysis patients were excluded. Measurements: Subjects underwent CMR with a 1.5-tesla whole-body magnetic resonance scanner
using a phased-array cardiac coil and retrospective vectorographic gating.
LV mass, LV ejection fraction (LVEF), LV end-systolic volume (LVESV), and LV
end-diastolic volume (LVEDV) were measured by CMR pre-KT and again 12 months
post-KT (N = 39), or 12 months later if still receiving dialysis (N = 43).
LV mass, LVESV, and LVEDV were indexed for height (m2.7) to
calculate left ventricular mass index (LVMI), left ventricular end-systolic
volume index (LVESVI), and left ventricular end-diastolic volume index
(LVEDVI), respectively. Serum total adiponectin and N-terminal proBNP
(NT-proBNP) concentrations were measured at baseline, 3 months, and 12
months. Methods: We performed a prospective 1:1 observational study comparing KT candidates
with ESKD either receiving a living donor organ (KT group) or waiting for a
deceased donor organ (dialysis group). Results: Left ventricular mass index change was −1.98 ± 5.5 and −0.36 ± 5.7
g/m2.7 for KT versus dialysis subjects (P =
.44). Left ventricular mass change was associated with systolic blood
pressure (SBP) (P = .0008) and average LV mass
(P = .0001). Left ventricular ejection fraction did not
improve (2.9 ± 6.6 vs 0.7 ± 4.9 %, P = .09), while LVESVI
and LVEDVI decreased more post-KT than with continued dialysis (−3.36 ± 5.6
vs −0.22 ± 4.4 mL/m2.7, P < .01 and −4.9 ±
8.5 vs −0.3 ± 9.2 mL/m2.7, P = .02). Both
adiponectin (−7.1 ± 11.3 vs −0.11 ± 7.9 µg/mL, P <
.0001) and NT-proBNP (−3811 ± 8130 vs 1665 ± 20013 pg/mL, P
< .0001) declined post-KT. Post-KT adiponectin correlated with NT-proBNP
(P = .001), but not estimated glomerular filtration
rate (eGFR) (P = .13). Change in adiponectin did not
correlate with change in LVEF in the KT group (Spearman ρ = 0.16,
P = .31) or dialysis group (Spearman ρ = 0.19,
P = .21). Limitations: Few biomarkers of cardiac function were measured to fully contextualize their
role during changing kidney function. Limited intrapatient biomarker
sampling and CMR measurements precluded constructing dose-response curves of
biomarkers to LV mass and function. The CMR timing in relation to dialysis
was not standardized. Conclusions: The LVESVI and LVEDVI but not LVMI or LVEF improve post-KT. LVMI and LVEF
change is independent of renal function and adiponectin. As adiponectin
correlates with NT-proBNP post-KT, improved renal function through KT
restores the normal heart-endocrine axis.
Collapse
Affiliation(s)
- G V Ramesh Prasad
- Division of Nephrology, St. Michael's Hospital, University of Toronto, ON, Canada
| | - Andrew T Yan
- Division of Cardiology, St. Michael's Hospital, University of Toronto, ON, Canada
| | - Michelle M Nash
- Renal Transplant Program, St. Michael's Hospital, Toronto, ON, Canada
| | - S Joseph Kim
- Division of Nephrology, Toronto General Hospital, University of Toronto, ON, Canada
| | - Ron Wald
- Division of Nephrology, St. Michael's Hospital, University of Toronto, ON, Canada
| | - Rachel Wald
- Division of Cardiology, Toronto General Hospital, University of Toronto, ON, Canada
| | - Charmaine Lok
- Division of Nephrology, Toronto General Hospital, University of Toronto, ON, Canada
| | - Lakshman Gunaratnam
- Division of Nephrology, London Health Sciences Centre, Western University, ON, Canada
| | - Gauri R Karur
- Division of Cardiology, St. Michael's Hospital, University of Toronto, ON, Canada
| | - Anish Kirpalani
- Department of Medical Imaging, St. Michael's Hospital, University of Toronto, ON, Canada
| | - Philip W Connelly
- Division of Endocrinology and Metabolism, St. Michael's Hospital, University of Toronto, ON, Canada
| |
Collapse
|
49
|
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
50
|
Connelly PW, Hanley AJ. Adiponectin, Adipokines, and the Need for Long-Term Human Studies With Comprehensive End Points. Arterioscler Thromb Vasc Biol 2018; 36:2136-2137. [PMID: 27784699 DOI: 10.1161/atvbaha.116.308402] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Philip W Connelly
- From the St. Michael's Hospital, Keenan Research Centre for Biomedical Science, Toronto, Ontario, Canada (P.W.C.); and Department of Nutritional Sciences, University of Toronto and Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, Ontario, Canada (A.J.H.).
| | - Anthony J Hanley
- From the St. Michael's Hospital, Keenan Research Centre for Biomedical Science, Toronto, Ontario, Canada (P.W.C.); and Department of Nutritional Sciences, University of Toronto and Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, Ontario, Canada (A.J.H.)
| |
Collapse
|