1
|
Kitsugi K, Chida T, Hanaoka T, Umemura M, Yamashita M, Ito J, Ohta K, Noritake H, Suda T, Kawata K. Elevated serum neprilysin levels in patients with chronic hepatitis C and metabolic dysfunction-associated steatotic liver disease: hepatic oxidative stress as an underlying mechanism. Mol Biol Rep 2024; 52:81. [PMID: 39722039 DOI: 10.1007/s11033-024-10152-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Neprilysin (NEP) is a metalloprotease that has become a therapeutic target for the treatment of heart failure and hypertension. However, the significance of NEP in chronic liver diseases has rarely been investigated. In this study, we investigated the serum NEP levels in patients with chronic liver disease and their relationship with clinical parameters. METHODS AND RESULTS Thirty-seven patients with chronic hepatitis C (CHC) who achieved sustained virologic response (SVR) after antiviral treatment and 73 patients with metabolic dysfunction-associated steatotic liver disease (MASLD) were enrolled. Serum neprilysin levels were measured using an enzyme-linked immunosorbent assay. The median NEP levels were 2.2 ng/mL in CHC and 4.1 ng/mL in MASLD, with the latter being significantly higher. Notably, in patients with MASLD, a significant correlation was observed between NEP and gamma-glutamyltransferase (GGT) levels at baseline. In contrast, there was no significant correlation between NEP levels and progression of liver fibrosis in either group. In the MASLD group, obesity and lifestyle diseases were significantly more prevalent, and the patients exhibited significantly higher NEP levels. In patients with CHC, NEP levels significantly decreased after SVR. NEP mRNA expression in liver tissues was significantly downregulated following SVR. Furthermore, a significant correlation was observed between the degree of NEP and GGT improvement. CONCLUSIONS Elevated NEP levels were observed in both CHC and MASLD groups. Considering the association between NEP levels and obesity, lifestyle diseases, and GGT levels, this suggests that oxidative stress may be involved in the elevation of NEP levels in patients with CHC and MASLD.
Collapse
Affiliation(s)
- Kensuke Kitsugi
- Department of Internal Medicine II, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Takeshi Chida
- Department of Internal Medicine II, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan.
- Department of Regional Medical Care Support, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan.
| | - Tomohiko Hanaoka
- Department of Internal Medicine II, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Masahiro Umemura
- Department of Internal Medicine II, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Maho Yamashita
- Department of Internal Medicine II, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Jun Ito
- Department of Internal Medicine II, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Kazuyoshi Ohta
- Department of Internal Medicine II, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Hidenao Noritake
- Department of Internal Medicine II, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Takafumi Suda
- Department of Internal Medicine II, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Kazuhito Kawata
- Department of Internal Medicine II, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| |
Collapse
|
2
|
Amdahl MB, Sundaram V, Reddy YNV. Obesity in Heart Failure with Reduced Ejection Fraction: Time to Address the Elephant in the Room. Heart Fail Clin 2024; 20:399-406. [PMID: 39216925 DOI: 10.1016/j.hfc.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Obesity has been long recognized as a risk factor for the development of heart failure, but recent evidence suggests obesity is more typically associated with heart failure with preserved ejection fraction as opposed to heart failure with reduced ejection fraction (HFrEF). Nevertheless, numerous studies have found that obesity modulates the presentation and progression of HFrEF and may contribute to the development of HFrEF in some patients. Although obesity has definite negative effects in HFrEF patients, the effects of intentional weight loss in HFrEF patients with obesity have been poorly studied.
Collapse
Affiliation(s)
- Matthew B Amdahl
- Department of Cardiovascular Diseases, Mayo Clinic, 200 1st Street Southwest, Rochester, MN 55905, USA
| | - Varun Sundaram
- Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Yogesh N V Reddy
- Department of Cardiovascular Diseases, Mayo Clinic, 200 1st Street Southwest, Rochester, MN 55905, USA; University Hospitals Medical Center, Cleveland, OH, USA.
| |
Collapse
|
3
|
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
|
4
|
Lembo M, Strisciuglio T, Fonderico C, Mancusi C, Izzo R, Trimarco V, Bellis A, Barbato E, Esposito G, Morisco C, Rubattu S. Obesity: the perfect storm for heart failure. ESC Heart Fail 2024; 11:1841-1860. [PMID: 38491741 PMCID: PMC11287355 DOI: 10.1002/ehf2.14641] [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: 07/14/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 03/18/2024] Open
Abstract
Obesity condition causes morphological and functional alterations involving the cardiovascular system. These can represent the substrates for different cardiovascular diseases, such as atrial fibrillation, coronary artery disease, sudden cardiac death, and heart failure (HF) with both preserved ejection fraction (EF) and reduced EF. Different pathogenetic mechanisms may help to explain the association between obesity and HF including left ventricular remodelling and epicardial fat accumulation, endothelial dysfunction, and coronary microvascular dysfunction. Multi-imaging modalities are required for appropriate recognition of subclinical systolic dysfunction typically associated with obesity, with echocardiography being the most cost-effective technique. Therapeutic approach in patients with obesity and HF is challenging, particularly regarding patients with preserved EF in which few strategies with high level of evidence are available. Weight loss is of extreme importance in patients with obesity and HF, being a primary therapeutic intervention. Sodium-glucose co-transporter-2 inhibitors have been recently introduced as a novel tool in the management of HF patients. The present review aims at analysing the most recent studies supporting pathogenesis, diagnosis, and management in patients with obesity and HF.
Collapse
Affiliation(s)
- Maria Lembo
- Department of Advanced Biochemical SciencesFederico II UniversityNaplesItaly
| | - Teresa Strisciuglio
- Department of Advanced Biochemical SciencesFederico II UniversityNaplesItaly
| | - Celeste Fonderico
- Department of Advanced Biochemical SciencesFederico II UniversityNaplesItaly
| | - Costantino Mancusi
- Department of Advanced Biochemical SciencesFederico II UniversityNaplesItaly
| | - Raffaele Izzo
- Department of Advanced Biochemical SciencesFederico II UniversityNaplesItaly
| | - Valentina Trimarco
- Department of Advanced Biochemical SciencesFederico II UniversityNaplesItaly
| | - Alessandro Bellis
- Emergenza Accettazione DepartmentAzienda Ospedaliera ‘Antonio Cardarelli’NaplesItaly
| | - Emanuele Barbato
- Department of Clinical and Molecular MedicineSapienza University of RomeRomeItaly
| | - Giovanni Esposito
- Department of Advanced Biochemical SciencesFederico II UniversityNaplesItaly
| | - Carmine Morisco
- Department of Advanced Biochemical SciencesFederico II UniversityNaplesItaly
| | - Speranza Rubattu
- Department of Clinical and Molecular MedicineSapienza University of RomeRomeItaly
- IRCCS NeuromedPozzilliItaly
| |
Collapse
|
5
|
Hoevelmann J, Markwirth P, Tokcan M, Haring B. What's new in heart failure? August-September 2024. Eur J Heart Fail 2024; 26:1665-1668. [PMID: 39331814 DOI: 10.1002/ejhf.3399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Affiliation(s)
- Julian Hoevelmann
- Department of Internal Medicine III, Saarland University Hospital, Homburg, Germany
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Philipp Markwirth
- Department of Internal Medicine III, Saarland University Hospital, Homburg, Germany
| | - Mert Tokcan
- Department of Internal Medicine III, Saarland University Hospital, Homburg, Germany
| | - Bernhard Haring
- Department of Internal Medicine III, Saarland University Hospital, Homburg, Germany
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
6
|
Babalola JA, Stracke A, Loeffler T, Schilcher I, Sideromenos S, Flunkert S, Neddens J, Lignell A, Prokesch M, Pazenboeck U, Strobl H, Tadic J, Leitinger G, Lass A, Hutter-Paier B, Hoefler G. Effect of astaxanthin in type-2 diabetes -induced APPxhQC transgenic and NTG mice. Mol Metab 2024; 85:101959. [PMID: 38763496 PMCID: PMC11153249 DOI: 10.1016/j.molmet.2024.101959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024] Open
Abstract
OBJECTIVES Aggregation and misfolding of amyloid beta (Aβ) and tau proteins, suggested to arise from post-translational modification processes, are thought to be the main cause of Alzheimer's disease (AD). Additionally, a plethora of evidence exists that links metabolic dysfunctions such as obesity, type 2 diabetes (T2D), and dyslipidemia to the pathogenesis of AD. We thus investigated the combinatory effect of T2D and human glutaminyl cyclase activity (pyroglutamylation), on the pathology of AD and whether astaxanthin (ASX) treatment ameliorates accompanying pathophysiological manifestations. METHODS Male transgenic AD mice, APPxhQC, expressing human APP751 with the Swedish and the London mutation and human glutaminyl cyclase (hQC) enzyme and their non-transgenic (NTG) littermates were used. Both APPxhQC and NTG mice were allocated to 3 groups, control, T2D-control, and T2D-ASX. Mice were fed control or high fat diet ± ASX for 13 weeks starting at an age of 11-12 months. High fat diet fed mice were further treated with streptozocin for T2D induction. Effects of genotype, T2D induction, and ASX treatment were evaluated by analysing glycemic readouts, lipid concentration, Aβ deposition, hippocampus-dependent cognitive function and nutrient sensing using immunosorbent assay, ELISA-based assays, western blotting, immunofluorescence staining, and behavioral testing via Morris water maze (MWM), respectively. RESULTS APPxhQC mice presented a higher glucose sensitivity compared to NTG mice. T2D-induced brain dysfunction was more severe in NTG compared to the APPxhQC mice. T2D induction impaired memory functions while increasing hepatic LC3B, ABCA1, and p65 levels in NTG mice. T2D induction resulted in a progressive shift of Aβ from the soluble to insoluble form in APPxhQC mice. ASX treatment reversed T2D-induced memory dysfunction in NTG mice and in parallel increased hepatic pAKT while decreasing p65 and increasing cerebral p-S6rp and p65 levels. ASX treatment reduced soluble Aβ38 and Aβ40 and insoluble Aβ40 levels in T2D-induced APPxhQC mice. CONCLUSIONS We demonstrate that T2D induction in APPxhQC mice poses additional risk for AD pathology as seen by increased Aβ deposition. Although ASX treatment reduced Aβ expression in T2D-induced APPxhQC mice and rescued T2D-induced memory impairment in NTG mice, ASX treatment alone may not be effective in cases of T2D comorbidity and AD.
Collapse
Affiliation(s)
| | - Anika Stracke
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Austria
| | | | | | - Spyridon Sideromenos
- QPS Austria GmbH, Grambach, Austria; Medical University of Vienna, Vienna, Austria
| | | | | | | | | | - Ute Pazenboeck
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Herbert Strobl
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Jelena Tadic
- Institute of Molecular Biosciences, University of Graz, Austria
| | - Gerd Leitinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Austria
| | | | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology Medical University of Graz, Graz, Austria.
| |
Collapse
|
7
|
Carrera-Alvarado G, Toldrá F, Mora L. Effect of thermal pretreatment and gastrointestinal digestion on the bioactivity of dry-cured ham bone enzymatic hydrolyzates. Food Res Int 2024; 188:114513. [PMID: 38823886 DOI: 10.1016/j.foodres.2024.114513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 06/03/2024]
Abstract
This study reports the effect of thermal pretreatment and the use of different commercial proteolytic enzymes (Protamex, Flavourzyme, Protana prime, and Alcalase) on the free amino acid content (FAA), peptide profile, and antioxidant, antidiabetic, antihypertensive, and anti-inflammatory potential (DPPH, FRAP, and ABTS assay, DPP-IV, ACE-I, and NEP inhibitory activities) of dry-cured ham bone hydrolyzates. The effect of in vitro digestion was also determined. Thermal pretreatment significantly increased the degree of hydrolysis, the FAA, and the DPP-IV and ACE-I inhibitory activities. The type of peptidase used was the most significant factor influencing antioxidant activity and neprilysin inhibitory activity. Protana prime hydrolyzates failed to inhibit DPP-IV and neprilysin enzymes and had low values of ACE-I inhibitory activity. After in vitro digestion, bioactivities kept constant in most cases or even increased in ACE-I inhibitory activity. Therefore, hydrolyzates from dry-cured ham bones could serve as a potential source of functional food ingredients for health benefits.
Collapse
Affiliation(s)
- Gisela Carrera-Alvarado
- Instituto de Agroquímica y Tecnología de Alimentos (CSIC), Avenue Agustín Escardino 7, 46980 Valencia, Paterna, Spain.
| | - Fidel Toldrá
- Instituto de Agroquímica y Tecnología de Alimentos (CSIC), Avenue Agustín Escardino 7, 46980 Valencia, Paterna, Spain.
| | - Leticia Mora
- Instituto de Agroquímica y Tecnología de Alimentos (CSIC), Avenue Agustín Escardino 7, 46980 Valencia, Paterna, Spain.
| |
Collapse
|
8
|
Yorek M. Combination therapy is it in the future for successfully treating peripheral diabetic neuropathy? Front Endocrinol (Lausanne) 2024; 15:1357859. [PMID: 38812811 PMCID: PMC11133577 DOI: 10.3389/fendo.2024.1357859] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/16/2024] [Indexed: 05/31/2024] Open
Abstract
In 2022, the Center for Disease Control and Prevention reported that 11.3% of the United States population, 37.3 million people, had diabetes and 38% of the population had prediabetes. A large American study conducted in 2021 and supported by many other studies, concluded that about 47% of diabetes patients have peripheral neuropathy and that diabetic neuropathy was present in 7.5% of patients at the time of diabetes diagnosis. In subjects deemed to be pre-diabetes and impaired glucose tolerance there was a wide range of prevalence estimates (interquartile range (IQR): 6%-34%), but most studies (72%) reported a prevalence of peripheral neuropathy ≥10%. There is no recognized treatment for diabetic peripheral neuropathy (DPN) other than good blood glucose control. Good glycemic control slows progression of DPN in patients with type 1 diabetes but for patients with type 2 diabetes it is less effective. With obesity and type 2 diabetes at epidemic levels the need of a treatment for DPN could not be more important. In this article I will first present background information on the "primary" mechanisms shown from pre-clinical studies to contribute to DPN and then discuss mono- and combination therapies that have demonstrated efficacy in animal studies and may have success when translated to human subjects. I like to compare the challenge of finding an effective treatment for DPN to the ongoing work being done to treat hypertension. Combination therapy is the recognized approach used to normalize blood pressure often requiring two, three or more drugs in addition to lifestyle modification to achieve the desired outcome. Hypertension, like DPN, is a progressive disease caused by multiple mechanisms. Therefore, it seems likely as well as logical that combination therapy combined with lifestyle adjustments will be required to successfully treat DPN.
Collapse
Affiliation(s)
- Mark Yorek
- Department of Internal Medicine, University of Iowa, Iowa City, IA, United States
- Department of Veterans Affairs Iowa City Health Care System, Iowa City, IA, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
9
|
Prickett TCR, Espiner EA, Pearson JF. Association of natriuretic peptides and receptor activity with cardio-metabolic health at middle age. Sci Rep 2024; 14:9919. [PMID: 38689031 PMCID: PMC11061163 DOI: 10.1038/s41598-024-60677-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024] Open
Abstract
Natriuretic peptides (NP) have multiple actions benefitting cardiovascular and metabolic health. Although many of these are mediated by Guanylyl Cyclase (GC) receptors NPR1 and NPR2, their role and relative importance in vivo is unclear. The intracellular mediator of NPR1 and NPR2, cGMP, circulates in plasma and can be used to examine relationships between receptor activity and tissue responses targeted by NPs. Plasma cGMP was measured in 348 participants previously recruited in a multidisciplinary community study (CHALICE) at age 50 years at a single centre. Associations between bio-active NPs and bio-inactive aminoterminal products with cGMP, and of cGMP with tissue response, were analysed using linear regression. Mediation of associations by NPs was assessed by Causal Mediation Analysis (CMA). ANP's contribution to cGMP far exceed those of other NPs. Modelling across three components (demographics, NPs and cardiovascular function) shows that ANP and CNP are independent and positive predictors of cGMP. Counter intuitively, findings from CMA imply that in specific tissues, NPR1 responds more to BNP stimulation than ANP. Collectively these findings align with longer tissue half-life of BNP, and direct further therapeutic interventions towards extending tissue activity of ANP and CNP.
Collapse
Affiliation(s)
- Timothy C R Prickett
- Departments of Medicine, University of Otago, Christchurch, PO Box 4345, Christchurch, 8140, New Zealand.
| | - Eric A Espiner
- Departments of Medicine, University of Otago, Christchurch, PO Box 4345, Christchurch, 8140, New Zealand
| | - John F Pearson
- Departments of Medicine, University of Otago, Christchurch, PO Box 4345, Christchurch, 8140, New Zealand
- Biostatistics and Computational Biology Unit, University of Otago, Christchurch, New Zealand
| |
Collapse
|
10
|
Wang S, Xiao Y, An X, Luo L, Gong K, Yu D. A comprehensive review of the literature on CD10: its function, clinical application, and prospects. Front Pharmacol 2024; 15:1336310. [PMID: 38389922 PMCID: PMC10881666 DOI: 10.3389/fphar.2024.1336310] [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: 11/14/2023] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
CD10, a zinc-dependent metalloprotease found on the cell surface, plays a pivotal role in an array of physiological and pathological processes including cardiovascular regulation, immune function, fetal development, pain response, oncogenesis, and aging. Recognized as a biomarker for hematopoietic and tissue stem cells, CD10 has garnered attention for its prognostic potential in the progression of leukemia and various solid tumors. Recent studies underscore its regulatory significance and therapeutic promise in combating Alzheimer's disease (AD), and it is noted for its protective role in preventing heart failure (HF), obesity, and type-2 diabetes. Furthermore, CD10/substance P interaction has also been shown to contribute to the pain signaling regulation and immunomodulation in diseases such as complex regional pain syndrome (CRPS) and osteoarthritis (OA). The emergence of COVID-19 has sparked interest in CD10's involvement in the disease's pathogenesis. Given its association with multiple disease states, CD10 is a prime therapeutic target; inhibitors targeting CD10 are now being advanced as therapeutic agents. This review compiles recent and earlier literature on CD10, elucidating its physicochemical attributes, tissue-specific expression, and molecular functions. Furthermore, it details the association of CD10 with various diseases and the clinical advancements of its inhibitors, providing a comprehensive overview of its growing significance in medical research.
Collapse
Affiliation(s)
- Shudong Wang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yinghui Xiao
- Public Research Platform, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xingna An
- Public Research Platform, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ling Luo
- Public Research Platform, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Kejian Gong
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dehai Yu
- Public Research Platform, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
11
|
Chaikijurajai T, Rincon-Choles H, Tang WHW. Natriuretic peptide testing strategies in heart failure: A 2023 update. Adv Clin Chem 2023; 118:155-203. [PMID: 38280805 DOI: 10.1016/bs.acc.2023.11.005] [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] [Indexed: 01/29/2024]
Abstract
Natriuretic peptides (NPs), including B-type natriuretic peptide (BNP) and N-terminal pro-BNP (NT-proBNP), have been recommended as standard biomarkers for diagnosing heart failure (HF), and one of the strongest risk predictors for mortality and HF hospitalization regardless of ejection fraction (EF) and etiology of HF. BNP is an active neurohormone opposing renin-angiotensin-aldosterone and sympathetic nervous system overactivated in HF, whereas NT-proBNP is an inactive prohormone released from cardiomyocytes in response to wall stress. Despite substantial advances in the development of guideline-directed medical therapy (GDMT) for HF with reduced EF, studies demonstrating direct benefits of NP-guided chronic HF therapy on mortality, HF hospitalization, and GDMT optimization have yielded conflicting results. However, accumulating evidence shows that achieving prespecified BNP or NT-proBNP target over time is significantly associated with favorable outcomes, suggesting that benefits of serially measured NPs may be limited to particular groups of HF patients, such as those with extreme levels of baseline BNP or NT-proBNP, which could represent severe phenotypes of HF associated with natriuretic peptide resistance or cardiorenal syndrome. Over the past decade, clinical utilization of BNP and NT-proBNP has been expanded, especially using serial NP measurements for guiding HF therapy, optimizing GDMT and identifying at-risk patients with HF phenotypes who may be minimally symptomatic or asymptomatic.
Collapse
Affiliation(s)
- Thanat Chaikijurajai
- Kaufman Center for Heart Failure Treatment and Recovery, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, United States; Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Hernan Rincon-Choles
- Department of Nephrology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, United States
| | - W H Wilson Tang
- Kaufman Center for Heart Failure Treatment and Recovery, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, United States.
| |
Collapse
|
12
|
Kjeldsen SA, Gluud LL, Werge MP, Pedersen JS, Bendtsen F, Alexiadou K, Tan T, Torekov SS, Iepsen EW, Jensen NJ, Richter MM, Goetze JP, Rungby J, Hartmann B, Holst JJ, Holst B, Holt J, Gustafsson F, Madsbad S, Svane MS, Bojsen-Møller KN, Wewer Albrechtsen NJ. Neprilysin activity is increased in metabolic dysfunction-associated steatotic liver disease and normalizes after bariatric surgery or GLP-1 therapy. iScience 2023; 26:108190. [PMID: 37953952 PMCID: PMC10638073 DOI: 10.1016/j.isci.2023.108190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/25/2023] [Accepted: 10/10/2023] [Indexed: 11/14/2023] Open
Abstract
Inhibitors of neprilysin improve glycemia in patients with heart failure and type 2 diabetes (T2D). The effect of weight loss by diet, surgery, or pharmacotherapy on neprilysin activity (NEPa) is unknown. We investigated circulating NEPa and neprilysin protein concentrations in obesity, T2D, metabolic dysfunction-associated steatotic liver disease (MASLD), and following bariatric surgery, or GLP-1-receptor-agonist therapy. NEPa, but not neprilysin protein, was enhanced in obesity, T2D, and MASLD. Notably, MASLD associated with NEPa independently of BMI and HbA1c. NEPa decreased after bariatric surgery with a concurrent increase in OGTT-stimulated GLP-1. Diet-induced weight loss did not affect NEPa, but individuals randomized to 52-week weight maintenance with liraglutide (1.2 mg/day) decreased NEPa, consistent with another study following 6-week liraglutide (3 mg/day). A 90-min GLP-1 infusion did not alter NEPa. Thus, MASLD may drive exaggerated NEPa, and lowered NEPa following bariatric surgery or liraglutide therapy may contribute to the reported improved cardiometabolic effects.
Collapse
Affiliation(s)
- Sasha A.S. Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, 2400 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Lise L. Gluud
- Gastro Unit, Copenhagen University Hospital Hvidovre, 2650 Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Mikkel P. Werge
- Gastro Unit, Copenhagen University Hospital Hvidovre, 2650 Hvidovre, Denmark
| | - Julie S. Pedersen
- Gastro Unit, Copenhagen University Hospital Hvidovre, 2650 Hvidovre, Denmark
| | - Flemming Bendtsen
- Gastro Unit, Copenhagen University Hospital Hvidovre, 2650 Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kleopatra Alexiadou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2BX, UK
| | - Tricia Tan
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2BX, UK
| | - Signe S. Torekov
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Eva W. Iepsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Nicole J. Jensen
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, 2400 Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
| | - Michael M. Richter
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, 2400 Copenhagen, Denmark
| | - Jens P. Goetze
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Jørgen Rungby
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, 2400 Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jens J. Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Birgitte Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Joachim Holt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Finn Gustafsson
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Cardiology, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Sten Madsbad
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Endocrinology, Copenhagen University Hospital - Hvidovre, 2650 Hvidovre, Denmark
| | - Maria S. Svane
- Gastro Unit, Copenhagen University Hospital Hvidovre, 2650 Hvidovre, Denmark
- Department of Endocrinology, Copenhagen University Hospital - Hvidovre, 2650 Hvidovre, Denmark
| | - Kirstine N. Bojsen-Møller
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Endocrinology, Copenhagen University Hospital - Hvidovre, 2650 Hvidovre, Denmark
| | - Nicolai J. Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, 2400 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
13
|
Amdahl MB, Sundaram V, Reddy YNV. Obesity in Heart Failure with Reduced Ejection Fraction: Time to Address the Elephant in the Room. Cardiol Clin 2023; 41:537-544. [PMID: 37743076 DOI: 10.1016/j.ccl.2023.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Obesity has been long recognized as a risk factor for the development of heart failure, but recent evidence suggests obesity is more typically associated with heart failure with preserved ejection fraction as opposed to heart failure with reduced ejection fraction (HFrEF). Nevertheless, numerous studies have found that obesity modulates the presentation and progression of HFrEF and may contribute to the development of HFrEF in some patients. Although obesity has definite negative effects in HFrEF patients, the effects of intentional weight loss in HFrEF patients with obesity have been poorly studied.
Collapse
Affiliation(s)
- Matthew B Amdahl
- Department of Cardiovascular Diseases, Mayo Clinic, 200 1st Street Southwest, Rochester, MN 55905, USA
| | - Varun Sundaram
- Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Yogesh N V Reddy
- Department of Cardiovascular Diseases, Mayo Clinic, 200 1st Street Southwest, Rochester, MN 55905, USA; University Hospitals Medical Center, Cleveland, OH, USA.
| |
Collapse
|
14
|
Esser N, Mongovin SM, Mundinger TO, Barrow BM, Zraika S. Neprilysin deficiency reduces hepatic gluconeogenesis in high fat-fed mice. Peptides 2023; 168:171076. [PMID: 37572792 PMCID: PMC10529503 DOI: 10.1016/j.peptides.2023.171076] [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: 04/28/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
Neprilysin is a peptidase that cleaves glucoregulatory peptides, including glucagon-like peptide-1 (GLP-1) and cholecystokinin (CCK). Some studies suggest that its inhibition in diabetes and/or obesity improves glycemia, and that this is associated with enhanced insulin secretion, glucose tolerance and insulin sensitivity. Whether reduced neprilysin activity also improves hepatic glucose metabolism has not been explored. We sought to determine whether genetic deletion of neprilysin suppresses hepatic glucose production (HGP) in high fat-fed mice. Nep+/+ and Nep-/- mice were fed high fat diet for 16 weeks, and then underwent a pyruvate tolerance test (PTT) to assess hepatic gluconeogenesis. Since glycogen breakdown in liver can also yield glucose, we assessed liver glycogen content in fasted and fed mice. In Nep-/- mice, glucose excursion during the PTT was reduced when compared to Nep+/+ mice. Further, liver glycogen levels were significantly greater in fasted but not fed Nep-/- versus Nep+/+ mice. Since gut-derived factors modulate HGP, we tested whether gut-selective inhibition of neprilysin could recapitulate the suppression of hepatic gluconeogenesis observed with whole-body inhibition, and this was indeed the case. Finally, the gut-derived neprilysin substrates, GLP-1 and CCK, are well-known to suppress HGP. Having previously demonstrated elevated plasma GLP-1 levels in Nep-/- mice, we now measured plasma CCK bioactivity and reveal an increase in Nep-/- versus Nep+/+ mice, suggesting GLP-1 and/or CCK may play a role in reducing HGP under conditions of neprilysin deficiency. In sum, neprilysin modulates hepatic gluconeogenesis and strategies to inhibit its activity may reduce HGP in type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Nathalie Esser
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States; Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States; Laboratory of Immunometabolism and Nutrition, GIGA-I3, CHU Liège, University of Liège, Liège, Belgium
| | - Stephen M Mongovin
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Thomas O Mundinger
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Breanne M Barrow
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Sakeneh Zraika
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States; Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States.
| |
Collapse
|
15
|
Preda A, Carbone F, Tirandi A, Montecucco F, Liberale L. Obesity phenotypes and cardiovascular risk: From pathophysiology to clinical management. Rev Endocr Metab Disord 2023; 24:901-919. [PMID: 37358728 PMCID: PMC10492705 DOI: 10.1007/s11154-023-09813-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/31/2023] [Indexed: 06/27/2023]
Abstract
Obesity epidemic reached the dimensions of a real global health crisis with more than one billion people worldwide living with obesity. Multiple obesity-related mechanisms cause structural, functional, humoral, and hemodynamic alterations with cardiovascular (CV) deleterious effects. A correct assessment of the cardiovascular risk in people with obesity is critical for reducing mortality and preserving quality of life. The correct identification of the obesity status remains difficult as recent evidence suggest that different phenotypes of obesity exist, each one associated with different degrees of CV risk. Diagnosis of obesity cannot depend only on anthropometric parameters but should include a precise assessment of the metabolic status. Recently, the World Heart Federation and World Obesity Federation provided an action plan for management of obesity-related CV risk and mortality, stressing for the instauration of comprehensive structured programs encompassing multidisciplinary teams. In this review we aim at providing an updated summary regarding the different obesity phenotypes, their specific effects on CV risk and differences in clinical management.
Collapse
Affiliation(s)
| | - Federico Carbone
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Amedeo Tirandi
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy.
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy.
| | - Luca Liberale
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| |
Collapse
|
16
|
Ichiki T, Cannone V, Scott CG, Iyer SR, Sangaralingham SJ, Bailey KR, Goetze JP, Tsuji Y, Rodeheffer RJ, Burnett JC. Sex-based differences in metabolic protection by the ANP genetic variant rs5068 in the general population. Am J Physiol Heart Circ Physiol 2023; 325:H545-H552. [PMID: 37417873 PMCID: PMC10538992 DOI: 10.1152/ajpheart.00321.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
Atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP) are produced in the heart and secreted into the circulation. As hormones, both peptides activate the guanylyl cyclase receptor A (GC-A), playing a role in blood pressure (BP) regulation. A significant role for ANP and BNP includes favorable actions in metabolic homeostasis. Sex-based high prevalence of risk factors for cardiovascular disease in males compared with females is well established, but sex-based differences on cardiometabolic protection have not been investigated in relation to ANP (NPPA) and BNP (NPPB) gene variants. We included 1,146 subjects in the general population from Olmsted County, Minnesota. Subjects were genotyped for the ANP gene variant rs5068 and BNP gene variant rs198389. Cardiometabolic parameters and medical records were reviewed. In the presence of the minor allele of rs5068, diastolic BP, creatinine, body mass index (BMI), waist measurement, insulin, and prevalence of obesity and metabolic syndrome were lower, whereas HDL was higher in males with only trends observed in females. We observed no associations of the minor allele with echocardiographic parameters in either males or females. Regarding rs198389 genotype, the minor allele was not associated with any BP, metabolic, renal, or echocardiographic parameters in either sex. In the general community, the minor allele of the ANP gene variant rs5068 is associated with a favorable metabolic phenotype in males. No associations were observed with the BNP gene variant rs198389. These studies support a protective role of the ANP pathway on metabolic function and underscore the importance of sex in relationship to natriuretic peptide responses.NEW & NOTEWORTHY Males are characterized by lower ANP and BNP with greater prevalence of cardiometabolic disease. The ANP genetic variant rs5068 was associated with less metabolic dysfunction in males, whereas no metabolic profile was related to the BNP genetic variant rs198389 in the general population. ANP may play a more biological role in metabolic homeostasis compared with BNP in the general population with greater physiological metabolic actions in males compared with females.
Collapse
Affiliation(s)
- Tomoko Ichiki
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Community and General Medicine, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Valentina Cannone
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Christopher G Scott
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States
| | - Seethalakshmi R Iyer
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Kent R Bailey
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States
| | - Jens P Goetze
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Yoshihisa Tsuji
- Department of Community and General Medicine, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Richard J Rodeheffer
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
17
|
Lin CY, Sung HY, Chen YJ, Yeh HI, Hou CJY, Tsai CT, Hung CL. Personalized Management for Heart Failure with Preserved Ejection Fraction. J Pers Med 2023; 13:jpm13050746. [PMID: 37240916 DOI: 10.3390/jpm13050746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous clinical syndrome with multiple underlying mechanisms and comorbidities that leads to a variety of clinical phenotypes. The identification and characterization of these phenotypes are essential for better understanding the precise pathophysiology of HFpEF, identifying appropriate treatment strategies, and improving patient outcomes. Despite accumulating data showing the potentiality of artificial intelligence (AI)-based phenotyping using clinical, biomarker, and imaging information from multiple dimensions in HFpEF management, contemporary guidelines and consensus do not incorporate these in daily practice. In the future, further studies are required to authenticate and substantiate these findings in order to establish a more standardized approach for clinical implementation.
Collapse
Affiliation(s)
- Chang-Yi Lin
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Road, Taipei 10449, Taiwan
| | - Heng-You Sung
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Road, Taipei 10449, Taiwan
| | - Ying-Ju Chen
- Telemedicine Center, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Hung-I Yeh
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Road, Taipei 10449, Taiwan
- Departments of Internal Medicine, Mackay Medical College, New Taipei City 25245, Taiwan
| | - Charles Jia-Yin Hou
- Departments of Internal Medicine, Mackay Medical College, New Taipei City 25245, Taiwan
| | - Cheng-Ting Tsai
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Road, Taipei 10449, Taiwan
- Mackay Junior College of Medicine, Nursing and Management, New Taipei City 25245, Taiwan
| | - Chung-Lieh Hung
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Road, Taipei 10449, Taiwan
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 25245, Taiwan
| |
Collapse
|
18
|
Hu J, Zhu H, Dai Y, Liu Y, Lu Y, Zhu S, Chen L, Zhang M, Jiang T, Peng H. Association between soluble neprilysin and diabetes: Findings from a prospective longitudinal study. Front Endocrinol (Lausanne) 2023; 14:1143590. [PMID: 37065764 PMCID: PMC10100928 DOI: 10.3389/fendo.2023.1143590] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/20/2023] [Indexed: 04/18/2023] Open
Abstract
Background The potential role of neprilysin (NEP) in glucose metabolism has been found by basic studies but lacks population evidence. The objective of this study was to examine the association between serum NEP and diabetes in Chinese adults. Methods In a prospective longitudinal cohort study - the Gusu cohort (n=2,286, mean age: 52 years, 61.5% females), the cross-sectional, longitudinal, and prospective associations between serum NEP and diabetes were systemically examined by logistic regression adjusting for conventional risk factors. Serum NEP was measured at baseline using commercial ELISA assays. Fasting glucose was repeatedly measured 4 years apart. Results The cross-sectional analysis found a positive association between serum NEP and fasting glucose at baseline (β=0.08, P=0.004 for log-transformed NEP). This association persisted after controlling for the dynamic risk profiles during follow-up (β=0.10, P=0.023 for log-transformed NEP). The prospective analysis found that a higher level of serum NEP at baseline was associated with a higher risk of diabetes during follow-up (OR=1.79, P=0.039 for log-transformed NEP). Conclusions Serum NEP was not only associated with prevalent diabetes but also predicted the future risk of diabetes development in Chinese adults, independent of many behavioral and metabolic factors. Serum NEP may be a predictor and even a new therapeutic target for diabetes. However, the casualty and mechanisms of NEP in the development of diabetes require further investigation.
Collapse
Affiliation(s)
- Junting Hu
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hanyun Zhu
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yunlang Dai
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Liu
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ying Lu
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Shasha Zhu
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Linan Chen
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Mingzhi Zhang
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Peng
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| |
Collapse
|
19
|
Schneider-Matyka D, Cybulska AM, Szkup M, Pilarczyk B, Panczyk M, Tomza-Marciniak A, Grochans E. Selenium as a predictor of metabolic syndrome in middle age women. Aging (Albany NY) 2023; 15:1734-1747. [PMID: 36947700 PMCID: PMC10085601 DOI: 10.18632/aging.204590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/04/2023] [Indexed: 03/24/2023]
Abstract
BACKGROUND Selenium plays an important role in metabolic homeostasis. It has been suggested that it may also affect the expression and activity of PPAR-γ. The aim of study was to analyze the relationships between these variables in the context of the health of women, for whom the risk of MetS increases with age. MATERIAL AND METHODS The study involved 390 women in middle age. The stages of study: a survey-based part; anthropometric measurements; analysis of biological material (blood) in terms of glycemia, triglyceride, HDL, and selenium levels, as well as genetic analysis of the PPAR-γ polymorphisms. RESULTS It was found that selenium may moderate the effect of the G allele of the PPAR-γ gene on the occurrence of elevated waist circumference (OR=1.030, 95%CI 1.005-1.057, p=0.020); and the effect of the C (OR=1.077, 95%CI 1.009-1.149, p=0.026) and the G alleles (OR=1.052, 95%CI 1.025-1.080, p<0.000) on the odds of elevated blood pressure. Women in whom HDL levels were not significantly reduced, had higher selenium levels (p=0.007). CONCLUSIONS 1. The effect of selenium on MetS and its components has not been demonstrated. 2. The effect of individual alleles of the PPAR-γ gene on MetS and its components was not demonstrated. 3. The concentration of selenium may affect waist circumference in carriers of the G allele, and arterial hypertension in carriers of the C and G alleles by affecting the expression of PPAR-γ. 4. Higher selenium concentrations increased the odds of higher HDL levels in the group of subjects meeting the MetS criteria.
Collapse
Affiliation(s)
- Daria Schneider-Matyka
- Department of Nursing, Pomeranian Medical University in Szczecin, Szczecin 71-210, Poland
| | - Anna Maria Cybulska
- Department of Nursing, Pomeranian Medical University in Szczecin, Szczecin 71-210, Poland
| | - Małgorzata Szkup
- Department of Nursing, Pomeranian Medical University in Szczecin, Szczecin 71-210, Poland
| | - Bogumiła Pilarczyk
- Department of Animal Reproduction Biotechnology and Environmental Hygiene, West Pomeranian University of Technology, Szczecin 71-217, Poland
| | - Mariusz Panczyk
- Department of Education and Research in Health Sciences, Faculty of Health Sciences, Medical University of Warsaw, Warsaw 00-581, Poland
| | - Agnieszka Tomza-Marciniak
- Department of Animal Reproduction Biotechnology and Environmental Hygiene, West Pomeranian University of Technology, Szczecin 71-217, Poland
| | - Elżbieta Grochans
- Department of Nursing, Pomeranian Medical University in Szczecin, Szczecin 71-210, Poland
| |
Collapse
|
20
|
Esser N, Mundinger TO, Barrow BM, Zraika S. Acute Inhibition of Intestinal Neprilysin Enhances Insulin Secretion via GLP-1 Receptor Signaling in Male Mice. Endocrinology 2023; 164:bqad055. [PMID: 36964914 PMCID: PMC10282919 DOI: 10.1210/endocr/bqad055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/15/2023] [Accepted: 03/22/2023] [Indexed: 03/26/2023]
Abstract
The peptidase neprilysin modulates glucose homeostasis by cleaving and inactivating insulinotropic peptides, including some produced in the intestine such as glucagon-like peptide-1 (GLP-1). Under diabetic conditions, systemic or islet-selective inhibition of neprilysin enhances beta-cell function through GLP-1 receptor (GLP-1R) signaling. While neprilysin is expressed in intestine, its local contribution to modulation of beta-cell function remains unknown. We sought to determine whether acute selective pharmacological inhibition of intestinal neprilysin enhanced glucose-stimulated insulin secretion under physiological conditions, and whether this effect was mediated through GLP-1R. Lean chow-fed Glp1r+/+ and Glp1r-/- mice received a single oral low dose of the neprilysin inhibitor thiorphan or vehicle. To confirm selective intestinal neprilysin inhibition, neprilysin activity in plasma and intestine (ileum and colon) was assessed 40 minutes after thiorphan or vehicle administration. In a separate cohort of mice, an oral glucose tolerance test was performed 30 minutes after thiorphan or vehicle administration to assess glucose-stimulated insulin secretion. Systemic active GLP-1 levels were measured in plasma collected 10 minutes after glucose administration. In both Glp1r+/+ and Glp1r-/- mice, thiorphan inhibited neprilysin activity in ileum and colon without altering plasma neprilysin activity or active GLP-1 levels. Further, thiorphan significantly increased insulin secretion in Glp1r+/+ mice, whereas it did not change insulin secretion in Glp1r-/- mice. In conclusion, under physiological conditions, acute pharmacological inhibition of intestinal neprilysin increases glucose-stimulated insulin secretion in a GLP-1R-dependent manner. Since intestinal neprilysin modulates beta-cell function, strategies to inhibit its activity specifically in the intestine may improve beta-cell dysfunction in type 2 diabetes.
Collapse
Affiliation(s)
- Nathalie Esser
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, CHU Liège, University of Liège, Liège, Belgium
| | - Thomas O Mundinger
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Breanne M Barrow
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Sakeneh Zraika
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
21
|
Ishihara S, Hiramitsu S, Kanaoka K, Taki M, Nakagawa H, Ueda T, Seno A, Nishida T, Onoue K, Soeda T, Ohtani T, Watanabe M, Kawakami R, Sakata Y, Kario K, Saito Y. New Conversion Formula Between B-Type Natriuretic Peptide and N-Terminal-Pro-B-Type Natriuretic Peptide - Analysis From a Multicenter Study. Circ J 2022; 86:2010-2018. [PMID: 35613887 DOI: 10.1253/circj.cj-22-0032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Although B-type natriuretic peptide (BNP) and N-terminal (NT)-proBNP are commonly used markers of heart failure, a simple conversion formula between these peptides has not yet been developed for clinical use. METHODS AND RESULTS A total of 9,394 samples were obtained from Nara Medical University, Jichi Medical University, and Osaka University. We randomly selected 70% for a derivation set to investigate a conversion formula from BNP to NT-proBNP using estimated glomerular filtration rate (eGFR) and body mass index (BMI); the remaining 30% was used as the internal validation set and we used a cohort study from Nara Medical University as an external validation set. Multivariate linear regression analysis revealed a new conversion formula: log NT-proBNP = 1.21 + 1.03 × log BNP - 0.009 × BMI - 0.007 × eGFR (r2=0.900, P<0.0001). The correlation coefficients between the actual and converted values of log NT-proBNP in the internal and external validation sets were 0.942 (P<0.0001) and 0.891 (P<0.0001), respectively. We applied this formula to samples obtained from patients administered with sacubitril/valsartan. After treatment initiation, NT-proBNP levels decreased and actual BNP levels increased. However, the calculated BNP levels decreased roughly parallel to the NT-proBNP levels. CONCLUSIONS This new and simple conversion formula of BNP and NT-proBNP with eGFR and BMI is potentially useful in clinical practice.
Collapse
Affiliation(s)
- Satomi Ishihara
- Department of Cardiovascular Medicine, Nara Medical University
| | | | - Koshiro Kanaoka
- Department of Cardiovascular Medicine, Nara Medical University
| | - Mizuri Taki
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine
| | | | - Tomoya Ueda
- Department of Cardiovascular Medicine, Nara Medical University
| | - Ayako Seno
- Department of Cardiovascular Medicine, Nara Medical University
| | - Taku Nishida
- Department of Cardiovascular Medicine, Nara Medical University
| | - Kenji Onoue
- Department of Cardiovascular Medicine, Nara Medical University
| | - Tsunenari Soeda
- Department of Cardiovascular Medicine, Nara Medical University
| | - Tomohito Ohtani
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Makoto Watanabe
- Department of Cardiovascular Medicine, Nara Medical University
| | - Rika Kawakami
- Department of Cardiovascular Medicine, Nara Medical University
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine
| | - Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University
| |
Collapse
|
22
|
Is bariatric surgery improving mitochondrial function in the renal cells of patients with obesity-induced kidney disease? Pharmacol Res 2022; 185:106488. [DOI: 10.1016/j.phrs.2022.106488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/22/2022]
|
23
|
Kagami K, Harada T, Ishii H, Obokata M. Key Phenotypes of Heart Failure with Preserved Ejection Fraction. Cardiol Clin 2022; 40:415-429. [DOI: 10.1016/j.ccl.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
24
|
Mahtani K, MMath BPBS, Brian Wang M, Barron A. Activation of GLP-1 receptor signalling by sacubitril/valsartan: Implications for patients with poor glycaemic control. Int J Cardiol 2022; 367:81-89. [DOI: 10.1016/j.ijcard.2022.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 07/24/2022] [Accepted: 08/04/2022] [Indexed: 12/11/2022]
|
25
|
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
|
26
|
Diabesity in Elderly Cardiovascular Disease Patients: Mechanisms and Regulators. Int J Mol Sci 2022; 23:ijms23147886. [PMID: 35887234 PMCID: PMC9318065 DOI: 10.3390/ijms23147886] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 12/04/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in the world. In 2019, 550 million people were suffering from CVD and 18 million of them died as a result. Most of them had associated risk factors such as high fasting glucose, which caused 134 million deaths, and obesity, which accounted for 5.02 million deaths. Diabesity, a combination of type 2 diabetes and obesity, contributes to cardiac, metabolic, inflammation and neurohumoral changes that determine cardiac dysfunction (diabesity-related cardiomyopathy). Epicardial adipose tissue (EAT) is distributed around the myocardium, promoting myocardial inflammation and fibrosis, and is associated with an increased risk of heart failure, particularly with preserved systolic function, atrial fibrillation and coronary atherosclerosis. In fact, several hypoglycaemic drugs have demonstrated a volume reduction of EAT and effects on its metabolic and inflammation profile. However, it is necessary to improve knowledge of the diabesity pathophysiologic mechanisms involved in the development and progression of cardiovascular diseases for comprehensive patient management including drugs to optimize glucometabolic control. This review presents the mechanisms of diabesity associated with cardiovascular disease and their therapeutic implications.
Collapse
|
27
|
Esser N, Schmidt C, Barrow BM, Cronic L, Hackney DJ, Mongovin SM, Hogan MF, Templin AT, Castillo JJ, Hull RL, Zraika S. Insulinotropic Effects of Neprilysin and/or Angiotensin Receptor Inhibition in Mice. Front Endocrinol (Lausanne) 2022; 13:888867. [PMID: 35733766 PMCID: PMC9207331 DOI: 10.3389/fendo.2022.888867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Treatment of heart failure with the angiotensin receptor-neprilysin inhibitor sacubitril/valsartan improved glycemic control in individuals with type 2 diabetes. The relative contribution of neprilysin inhibition versus angiotensin II receptor antagonism to this glycemic benefit remains unknown. Thus, we sought to determine the relative effects of the neprilysin inhibitor sacubitril versus the angiotensin II receptor blocker valsartan on beta-cell function and glucose homeostasis in a mouse model of reduced first-phase insulin secretion, and whether any beneficial effects are additive/synergistic when combined in sacubitril/valsartan. High fat-fed C57BL/6J mice treated with low-dose streptozotocin (or vehicle) were followed for eight weeks on high fat diet alone or supplemented with sacubitril, valsartan or sacubitril/valsartan. Body weight and fed glucose levels were assessed weekly. At the end of the treatment period, insulin release in response to intravenous glucose, insulin sensitivity, and beta-cell mass were determined. Sacubitril and valsartan, but not sacubitril/valsartan, lowered fasting and fed glucose levels and increased insulin release in diabetic mice. None of the drugs altered insulin sensitivity or beta-cell mass, but all reduced body weight gain. Effects of the drugs on insulin release were reproduced in angiotensin II-treated islets from lean C57BL/6J mice, suggesting the insulin response to each of the drugs is due to a direct effect on islets and mechanisms therein. In summary, sacubitril and valsartan each exert beneficial insulinotropic, glycemic and weight-reducing effects in obese and/or diabetic mice when administered alone; however, when combined, mechanisms within the islet contribute to their inability to enhance insulin release.
Collapse
Affiliation(s)
- Nathalie Esser
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
- Laboratory of Immunometabolism and Nutrition, GIGA Infection, Immunity and Inflammation, University of Liège, Liège, Belgium
| | - Christine Schmidt
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Breanne M. Barrow
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Laura Cronic
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Daryl J. Hackney
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Stephen M. Mongovin
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Meghan F. Hogan
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Andrew T. Templin
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Joseph J. Castillo
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Rebecca L. Hull
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Sakeneh Zraika
- Research Service, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
28
|
Abstract
The development of pulmonary hypertension (PH) is common and has adverse prognostic implications in patients with heart failure due to left heart disease (LHD), and thus far, there are no known treatments specifically for PH-LHD, also known as group 2 PH. Diagnostic thresholds for PH-LHD, and clinical classification of PH-LHD phenotypes, continue to evolve and, therefore, present a challenge for basic and translational scientists actively investigating PH-LHD in the preclinical setting. Furthermore, the pathobiology of PH-LHD is not well understood, although pulmonary vascular remodeling is thought to result from (1) increased wall stress due to increased left atrial pressures; (2) hemodynamic congestion-induced decreased shear stress in the pulmonary vascular bed; (3) comorbidity-induced endothelial dysfunction with direct injury to the pulmonary microvasculature; and (4) superimposed pulmonary arterial hypertension risk factors. To ultimately be able to modify disease, either by prevention or treatment, a better understanding of the various drivers of PH-LHD, including endothelial dysfunction, abnormalities in vascular tone, platelet aggregation, inflammation, adipocytokines, and systemic complications (including splanchnic congestion and lymphatic dysfunction) must be further investigated. Here, we review the diagnostic criteria and various hemodynamic phenotypes of PH-LHD, the potential biological mechanisms underlying this disorder, and pressing questions yet to be answered about the pathobiology of PH-LHD.
Collapse
Affiliation(s)
- Jessica H Huston
- Division of Cardiology, Department of Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA (J.H.H.)
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.)
| |
Collapse
|
29
|
Westmark CJ, Filon MJ, Maina P, Steinberg LI, Ikonomidou C, Westmark PR. Effects of Soy-Based Infant Formula on Weight Gain and Neurodevelopment in an Autism Mouse Model. Cells 2022; 11:1350. [PMID: 35456030 PMCID: PMC9025435 DOI: 10.3390/cells11081350] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 02/01/2023] Open
Abstract
Mice fed soy-based diets exhibit increased weight gain compared to mice fed casein-based diets, and the effects are more pronounced in a model of fragile X syndrome (FXS; Fmr1KO). FXS is a neurodevelopmental disability characterized by intellectual impairment, seizures, autistic behavior, anxiety, and obesity. Here, we analyzed body weight as a function of mouse age, diet, and genotype to determine the effect of diet (soy, casein, and grain-based) on weight gain. We also assessed plasma protein biomarker expression and behavior in response to diet. Juvenile Fmr1KO mice fed a soy protein-based rodent chow throughout gestation and postnatal development exhibit increased weight gain compared to mice fed a casein-based purified ingredient diet or grain-based, low phytoestrogen chow. Adolescent and adult Fmr1KO mice fed a soy-based infant formula diet exhibited increased weight gain compared to reference diets. Increased body mass was due to increased lean mass. Wild-type male mice fed soy-based infant formula exhibited increased learning in a passive avoidance paradigm, and Fmr1KO male mice had a deficit in nest building. Thus, at the systems level, consumption of soy-based diets increases weight gain and affects behavior. At the molecular level, a soy-based infant formula diet was associated with altered expression of numerous plasma proteins, including the adipose hormone leptin and the β-amyloid degrading enzyme neprilysin. In conclusion, single-source, soy-based diets may contribute to the development of obesity and the exacerbation of neurological phenotypes in developmental disabilities, such as FXS.
Collapse
Affiliation(s)
- Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
- Molecular Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, USA
| | - Mikolaj J. Filon
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
- Undergraduate Research Program, University of Wisconsin, Madison, WI 53706, USA
| | - Patricia Maina
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
- Molecular Environmental Toxicology Summer Research Opportunities Program, University of Wisconsin, Madison, WI 53706, USA
| | - Lauren I. Steinberg
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
- Undergraduate Research Program, University of Wisconsin, Madison, WI 53706, USA
| | - Chrysanthy Ikonomidou
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
| | - Pamela R. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
| |
Collapse
|
30
|
Remodeling and Fibrosis of the Cardiac Muscle in the Course of Obesity-Pathogenesis and Involvement of the Extracellular Matrix. Int J Mol Sci 2022; 23:ijms23084195. [PMID: 35457013 PMCID: PMC9032681 DOI: 10.3390/ijms23084195] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/09/2022] [Indexed: 12/16/2022] Open
Abstract
Obesity is a growing epidemiological problem, as two-thirds of the adult population are carrying excess weight. It is a risk factor for the development of cardiovascular diseases (hypertension, ischemic heart disease, myocardial infarct, and atrial fibrillation). It has also been shown that chronic obesity in people may be a cause for the development of heart failure with preserved ejection fraction (HFpEF), whose components include cellular hypertrophy, left ventricular diastolic dysfunction, and increased extracellular collagen deposition. Several animal models with induced obesity, via the administration of a high-fat diet, also developed increased heart fibrosis as a result of extracellular collagen accumulation. Excessive collagen deposition in the extracellular matrix (ECM) in the course of obesity may increase the stiffness of the myocardium and thereby deteriorate the heart diastolic function and facilitate the occurrence of HFpEF. In this review, we include a rationale for that process, including a discussion about possible putative factors (such as increased renin–angiotensin–aldosterone activity, sympathetic overdrive, hemodynamic alterations, hypoadiponectinemia, hyperleptinemia, and concomitant heart diseases). To address the topic clearly, we include a description of the fundamentals of ECM turnover, as well as a summary of studies assessing collagen deposition in obese individuals.
Collapse
|
31
|
CANDER S, ÖZ GÜL Ö. Investigation of serum neprilysin levels in overweight and normal weight young women. TURKISH JOURNAL OF INTERNAL MEDICINE 2022. [DOI: 10.46310/tjim.1070422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
32
|
Conte M, Petraglia L, Poggio P, Valerio V, Cabaro S, Campana P, Comentale G, Attena E, Russo V, Pilato E, Formisano P, Leosco D, Parisi V. Inflammation and Cardiovascular Diseases in the Elderly: The Role of Epicardial Adipose Tissue. Front Med (Lausanne) 2022; 9:844266. [PMID: 35242789 PMCID: PMC8887867 DOI: 10.3389/fmed.2022.844266] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/13/2022] [Indexed: 01/08/2023] Open
Abstract
Human aging is a complex phenomenon characterized by a wide spectrum of biological changes which impact on behavioral and social aspects. Age-related changes are accompanied by a decline in biological function and increased vulnerability leading to frailty, thereby advanced age is identified among the major risk factors of the main chronic human diseases. Aging is characterized by a state of chronic low-grade inflammation, also referred as inflammaging. It recognizes a multifactorial pathogenesis with a prominent role of the innate immune system activation, resulting in tissue degeneration and contributing to adverse outcomes. It is widely recognized that inflammation plays a central role in the development and progression of numerous chronic and cardiovascular diseases. In particular, low-grade inflammation, through an increased risk of atherosclerosis and insulin resistance, promote cardiovascular diseases in the elderly. Low-grade inflammation is also promoted by visceral adiposity, whose accumulation is paralleled by an increased inflammatory status. Aging is associated to increase in epicardial adipose tissue (EAT), the visceral fat depot of the heart. Structural and functional changes in EAT have been shown to be associated with several heart diseases, including coronary artery disease, aortic stenosis, atrial fibrillation, and heart failure. EAT increase is associated with a greater production and secretion of pro-inflammatory mediators and neuro-hormones, so that thickened EAT can pathologically influence, in a paracrine and vasocrine manner, the structure and function of the heart and is associated to a worse cardiovascular outcome. In this review, we will discuss the evidence underlying the interplay between inflammaging, EAT accumulation and cardiovascular diseases. We will examine and discuss the importance of EAT quantification, its characteristics and changes with age and its clinical implication.
Collapse
Affiliation(s)
- Maddalena Conte
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Casa di Cura San Michele, Maddaloni, Italy
| | - Laura Petraglia
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | | | | | - Serena Cabaro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Pasquale Campana
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Giuseppe Comentale
- Department of Advanced Biomedical Science, University of Naples Federico II, Naples, Italy
| | - Emilio Attena
- Department of Cardiology, Monaldi Hospital, Naples, Italy
| | - Vincenzo Russo
- Department of Medical Translational Sciences, Monaldi Hospital, University of Campania Luigi Vanvitelli, Campania, Italy
| | - Emanuele Pilato
- Department of Advanced Biomedical Science, University of Naples Federico II, Naples, Italy
| | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Dario Leosco
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Valentina Parisi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
33
|
Esser N, Mongovin SM, Parilla J, Barrow BM, Mundinger TO, Fountaine BS, Larmore MJ, Castillo JJ, Akter R, Hull RL, Zraika S. Neprilysin inhibition improves intravenous but not oral glucose-mediated insulin secretion via GLP-1R signaling in mice with β-cell dysfunction. Am J Physiol Endocrinol Metab 2022; 322:E307-E318. [PMID: 35128957 PMCID: PMC8917916 DOI: 10.1152/ajpendo.00234.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Type 2 diabetes is associated with the upregulation of neprilysin, a peptidase capable of cleaving glucoregulatory peptides such as glucagon-like peptide-1 (GLP-1). In humans, use of the neprilysin inhibitor sacubitril in combination with an angiotensin II receptor blocker was associated with increased plasma GLP-1 levels and improved glycemic control. Whether neprilysin inhibition per se is mediating these effects remains unknown. We sought to determine whether pharmacological neprilysin inhibition on its own confers beneficial effects on glycemic status and β-cell function in a mouse model of reduced insulin secretion, and whether any such effects are dependent on GLP-1 receptor (GLP-1R) signaling. High-fat-fed male wild-type (Glp1r+/+) and GLP-1R knockout (Glp1r-/-) mice were treated with low-dose streptozotocin (STZ) to recapitulate type 2 diabetes-associated β-cell dysfunction, or vehicle as control. Mice were continued on high-fat diet alone or supplemented with the neprilysin inhibitor sacubitril for 8 wk. At the end of the study period, β-cell function was assessed by oral or intravenous glucose-tolerance test. Fasting and fed glucose were significantly lower in wild-type mice treated with sacubitril, although active GLP-1 levels and insulin secretion during oral glucose challenge were unchanged. In contrast, insulin secretion in response to intravenous glucose was significantly enhanced in sacubitril-treated wild-type mice, and this effect was blunted in Glp1r-/- mice. Similarly, sacubitril enhanced insulin secretion in vitro in islets from STZ-treated Glp1r+/+ but not Glp1r-/- mice. Together, our data suggest the insulinotropic effects of pharmacological neprilysin inhibition in a mouse model of β-cell dysfunction are mediated via intra-islet GLP-1R signaling.NEW & NOTEWORTHY The neprilysin inhibitor, sacubitril, improves glycemic status in a mouse model of reduced insulin secretion. Sacubitril enhances intravenous but not oral glucose-mediated insulin secretion. The increased glucose-mediated insulin secretion is GLP-1 receptor-dependent. Neprilysin inhibition does not raise postprandial circulating active GLP-1 levels.
Collapse
Affiliation(s)
- Nathalie Esser
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
| | | | - Jacqueline Parilla
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| | - Breanne M Barrow
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
| | - Thomas O Mundinger
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| | | | - Megan J Larmore
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Joseph J Castillo
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| | - Rehana Akter
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| | - Rebecca L Hull
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| | - Sakeneh Zraika
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
34
|
Walker JM, Kazempour Dehkordi S, Fracassi A, Vanschoiack A, Pavenko A, Taglialatela G, Woltjer R, Richardson TE, Zare H, Orr ME. Differential protein expression in the hippocampi of resilient individuals identified by digital spatial profiling. Acta Neuropathol Commun 2022; 10:23. [PMID: 35164877 PMCID: PMC8842950 DOI: 10.1186/s40478-022-01324-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/30/2022] [Indexed: 02/04/2023] Open
Abstract
Clinical symptoms correlate with underlying neurodegenerative changes in the vast majority of people. However, an intriguing group of individuals demonstrate neuropathologic changes consistent with Alzheimer disease (AD) yet remain cognitively normal (termed "resilient"). Previous studies have reported less overall neuronal loss, less gliosis, and fewer comorbidities in these individuals. Herein, NanoString GeoMx™ Digital Spatial Profiler (DSP) technology was utilized to investigate protein expression differences comparing individuals with dementia and AD neuropathologic change to resilient individuals. DSP allows for spatial analysis of protein expression in multiple regions of interest (ROIs) on formalin-fixed paraffin-embedded sections. ROIs in this analysis were hippocampal neurofibrillary tangle (NFT)-bearing neurons, non-NFT-bearing neurons, and their immediate neuronal microenvironments. Analyses of 86 proteins associated with CNS cell-typing or known neurodegenerative changes in 168 ROIs from 14 individuals identified 11 proteins displaying differential expression in NFT-bearing neurons of the resilient when compared to the demented (including APP, IDH1, CD68, GFAP, SYP and Histone H3). In addition, IDH1, CD68, and SYP were differentially expressed in the environment of NFT-bearing neurons when comparing resilient to demented. IDH1 (which is upregulated under energetic and oxidative stress) and PINK1 (which is upregulated in response to mitochondrial dysfunction and oxidative stress) both displayed lower expression in the environment of NFT-bearing neurons in the resilient. Therefore, the resilient display less evidence of energetic and oxidative stress. Synaptophysin (SYP) was increased in the resilient, which likely indicates better maintenance of synapses and synaptic connections. Furthermore, neurofilament light chain (NEFL) and ubiquitin c-terminal hydrolase (Park5) were higher in the resilient in the environment of NFTs. These differences all suggest healthier intact axons, dendrites and synapses in the resilient. In conclusion, resilient individuals display protein expression patterns suggestive of an environment containing less energetic and oxidative stress, which in turn results in maintenance of neurons and their synaptic connections.
Collapse
Affiliation(s)
- Jamie M. Walker
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center, San Antonio, TX USA
- Department of Pathology, Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, 7703 Floyd Curl Dr., MC 8070, San Antonio, TX 78229-3900 USA
| | - Shiva Kazempour Dehkordi
- Department of Pathology, Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, 7703 Floyd Curl Dr., MC 8070, San Antonio, TX 78229-3900 USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX USA
| | - Anna Fracassi
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB, Galveston, TX USA
| | | | | | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, UTMB, Galveston, TX USA
| | - Randall Woltjer
- Department of Pathology & Laboratory Medicine, Oregon Health and Science University, Portland, OR USA
| | - Timothy E. Richardson
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center, San Antonio, TX USA
- Department of Pathology, Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, 7703 Floyd Curl Dr., MC 8070, San Antonio, TX 78229-3900 USA
| | - Habil Zare
- Department of Pathology, Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center, 7703 Floyd Curl Dr., MC 8070, San Antonio, TX 78229-3900 USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX USA
| | - Miranda E. Orr
- Section of Gerontology and Geriatric Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Salisbury VA Medical Center, Salisbury, NC 28144 USA
- Department of Internal Medicine, Wake Forest School of Medicine, 575 Patterson Ave, Winston-Salem, NC 27101 USA
| |
Collapse
|
35
|
Gul OO, Sisman P, Cander S, Sarandol E, Ersoy C, Erturk E. PLASMA NEPRILYSIN LEVELS IN PATIENTS WITH POLYCYSTIC OVARY SYNDROME. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2022; 18:35-39. [PMID: 35975244 PMCID: PMC9365417 DOI: 10.4183/aeb.2022.35] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
CONTEXT Insulin resistance has been detected in a majority of patients with polycystic ovary syndrome (PCOS). Elevated neprilysin levels are associated with insulin resistance. OBJECTIVE The present study aims to investigate plasma neprilysin and its relationship with endocrine and metabolic characteristics in patients with PCOS. SUBJECTS AND METHODS Thirty-five premenopausal PCOS patients and 35 healthy volunteers of similar age were included in the study. Demographic characteristics, biochemical and hormonal findings and also plasma neprilysin levels were determined in these patients and healthy controls. RESULTS In our study, HOMA-IR values were significantly higher in PCOS patients (3.3 ± 1.8) compared with the controls [(1.6 ± 1), p<0.01]. Plasma neprilysin levels were significantly higher in the PCOS group compared to the control group (1502.1 ± 1641.2 vs. 764.6 ± 562.6 pg/mL). There was no difference in plasma neprilysin levels when PCOS patients were classified as overweight-obesity (BMI≥25kg/m2) or non-obesity (BMI<25kg/m2). CONCLUSION Our findings revealed significantly higher levels for plasma neprilysin and HOMA-IR values in PCOS patients when compared to controls. No significant differences were noted between obese PCOS patients and non-obese PCOS patients in terms of plasma neprilysin levels.
Collapse
Affiliation(s)
- O. Oz Gul
- Uludag University Medical School, Department of Endocrinology and Metabolism, Bursa, Turkey
| | - P. Sisman
- Medicana Hospital, Endocrinology and Metabolism Clinic, Bursa, Turkey
| | - S. Cander
- Uludag University Medical School, Department of Endocrinology and Metabolism, Bursa, Turkey
| | - E. Sarandol
- Uludag University Medical School, Department of Biochemistry, Bursa, Turkey
| | - C. Ersoy
- Uludag University Medical School, Department of Endocrinology and Metabolism, Bursa, Turkey
| | - E. Erturk
- Uludag University Medical School, Department of Endocrinology and Metabolism, Bursa, Turkey
| |
Collapse
|
36
|
Lin W, Liu Z, Li L, Zhou Z, Ma S, He F. Effects of Shugan Quzhi Capsule in treating different metabolic diseases based on network pharmacology and molecular docking. 2021 IEEE INTERNATIONAL CONFERENCE ON BIOINFORMATICS AND BIOMEDICINE (BIBM) 2021:3832-3839. [DOI: 10.1109/bibm52615.2021.9669290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
37
|
Obokata M, Reddy YNV, Melenovsky V, Sorimachi H, Jarolim P, Borlaug BA. Uncoupling between intravascular and distending pressures leads to underestimation of circulatory congestion in obesity. Eur J Heart Fail 2021; 24:353-361. [PMID: 34755429 DOI: 10.1002/ejhf.2377] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/15/2021] [Accepted: 11/01/2021] [Indexed: 12/20/2022] Open
Abstract
AIMS Patients with obesity frequently present with dyspnoea. Biomarkers that reflect wall stress are often used to evaluate circulatory congestion and help determine whether dyspnoea is of cardiac causes. Patients with obesity display greater external restraint on the heart, which may alter relationships between intravascular pressures and stress markers. METHODS AND RESULTS Subjects with unexplained dyspnoea (n = 212) underwent cardiac catheterization with simultaneous echocardiography. Blood sampling was performed in a subset (n = 58). Relationships between echocardiographic and blood biomarkers of circulatory congestion and directly-measured haemodynamics were compared between participants with severe obesity [body mass index (BMI) ≥35 kg/m2 , Group B) and those without (BMI <35 kg/m2 , Group A). Circulatory congestion was assessed by pulmonary capillary wedge pressure (PCWP), and vascular distending pressure was assessed by left ventricular transmural pressure (LVTMP). As compared to Group A, participants in Group B displayed higher PCWP relative to N-terminal pro-B-type natriuretic peptide, mid-regional pro-atrial natriuretic peptide (MR-proANP), troponin T, and growth differentiation factor-15 (all p < 0.01). In contrast, the relationships between LVTMP and the biomarkers were superimposable. Echocardiographic biomarkers revealed the same pattern: PCWP was higher for any E/e' ratio in Group B compared to Group A, but the relationship between LVTMP and E/e' was similar. In contrast, levels of C-terminal pro-endothelin-1 and MR-proADM were more robustly correlated with PCWP (r = 0.67 and r = 0.62, both p < 0.0001), with no differential relationship based upon BMI. CONCLUSIONS Non-invasive haemodynamic markers underestimate circulatory congestion in patients with obesity, an effect that appears related to uncoupling between cardiac wall stress and intravascular pressures. This may lead to systematic under-recognition of congestion in patients with obesity.
Collapse
Affiliation(s)
- Masaru Obokata
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.,Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yogesh N V Reddy
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Vojtech Melenovsky
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.,Institute for Clinical and Experimental Medicine-IKEM, Prague, Czech Republic
| | - Hidemi Sorimachi
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Petr Jarolim
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Barry A Borlaug
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
38
|
Liu X, Patel KP, Zheng H. Role of Renal Sympathetic Nerves in GLP-1 (Glucagon-Like Peptide-1) Receptor Agonist Exendin-4-Mediated Diuresis and Natriuresis in Diet-Induced Obese Rats. J Am Heart Assoc 2021; 10:e022542. [PMID: 34713714 PMCID: PMC8751817 DOI: 10.1161/jaha.121.022542] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background The gut‐derived hormone GLP‐1 (glucagon‐like peptide‐1) exerts beneficial effects against established risk factors for chronic kidney disease. GLP‐1 influences renal function by stimulating diuresis and natriuresis and thus lowering arterial blood pressure. The role of the sympathetic nervous system has been implicated as an important link between obesity with elevated arterial pressure and chronic kidney disease. The primary aim of this study was to determine the contribution of renal sympathetic nerves on intrapelvic GLP‐1‐mediated diuresis and natriuresis in high‐fat diet (HFD)‐induced obese rats. Methods and Results Obesity was induced in rats by HFD for 12 weeks, followed by either surgical bilateral renal denervation or chronic subcutaneous endopeptidase neprilysin inhibition by sacubitril for a week. Diuretic and natriuretic responses to intrapelvic administration of the GLP‐1R (GLP‐1 receptor) agonist exendin‐4 were monitored in anesthetized control and HFD rats. Renal GLP‐1R expression and neprilysin expression and activity were measured. The effects of norepinephrine on the expression of GLP‐1R and neprilysin in kidney epithelial LLC‐PK1 cells were also examined. We found that diuretic and natriuretic responses to exendin‐4 were significantly reduced in the HFD obese rats compared with the control rats (cumulative urine flow at 40 minutes, 387±32 versus 650±65 µL/gkw; cumulative sodium excretion at 40 minutes, 42±5 versus 75±10 µEq/gkw, P<0.05). These responses in the HFD rats were restored after ablation of renal nerves (cumulative urine flow at 40 minutes, 625±62 versus 387±32 µL/gkw; cumulative sodium excretion at 40 minutes, 70±9 versus 42±5 µEq/gkw, P<0.05). Renal denervation induced significant reductions in arterial pressure and heart rate responses to intrapelvic GLP‐1 in the HFD rats. Renal denervation also significantly increased the GLP‐1R expression and reduced neprilysin expression and activity in renal tissues from the HFD rats. Chronic subcutaneous neprilysin inhibition by sacubitril increased GLP‐1–induced diuretic and natriuretic effects in the HFD rats. Finally, exposure of the renal epithelial cells to norepinephrine in vitro led to downregulation of GLP‐1R expression but upregulation of neprilysin expression and activity. Conclusions These results suggest that renal sympathetic nerve activation contributes to the blunted diuretic and natriuretic effects of GLP‐1 in HFD obese rats. This study provides significant novel insight into the potential renal nerve–neprilysin–GLP‐1 pathway involved in renal dysfunction during obesity that leads to hypertension.
Collapse
Affiliation(s)
- Xuefei Liu
- Division of Basic Biomedical Sciences Sanford School of Medicine of the University of South Dakota Vermillion SD
| | - Kaushik P Patel
- Department of Cellular and Integrative Physiology University of Nebraska Medical Center Omaha NE
| | - Hong Zheng
- Division of Basic Biomedical Sciences Sanford School of Medicine of the University of South Dakota Vermillion SD
| |
Collapse
|
39
|
van de Bovenkamp AA, Enait V, de Man FS, Oosterveer FTP, Bogaard HJ, Vonk Noordegraaf A, van Rossum AC, Handoko ML. Validation of the 2016 ASE/EACVI Guideline for Diastolic Dysfunction in Patients With Unexplained Dyspnea and a Preserved Left Ventricular Ejection Fraction. J Am Heart Assoc 2021; 10:e021165. [PMID: 34476984 PMCID: PMC8649534 DOI: 10.1161/jaha.121.021165] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Echocardiography is considered the cornerstone of the diagnostic workup of heart failure with preserved ejection fraction. Thus far, validation of the 2016 American Society of Echocardiography/European Association of Cardiovascular Imaging (ASE/EACVI) echo‐algorithm for evaluation of diastolic (dys)function in a patient suspected of heart failure with preserved ejection fraction has been limited. Methods and Results The diagnostic performance of the 2016 ASE/EACVI algorithm was assessed in 204 patients evaluated for unexplained dyspnea or pulmonary hypertension with echocardiogram and right heart catheterization. Invasively measured pulmonary capillary wedge pressure (PCWP) was used as the gold standard. In addition, the diagnostic performance of H2FPEF score and NT‐proBNP (N‐terminal pro‐B‐type natriuretic peptide) were evaluated. There was a poor correlation between indexed left atrial volume, E/e′ (septal and average) or early mitral inflow (E), and PCWP (r=0.25–0.30, P values all <0.01). No correlation was found in our cohort between e′ (septal or lateral) or tricuspid valve regurgitation and PCWP. The correlation between diastolic function grades of the ASE/EACVI algorithm and PCWP was poor (r=0.17, P<0.05). The ASE/EACVI algorithm had a sensitivity and specificity of 35% and 87%, respectively; an accuracy of 67% and an area under the curve of 0.56. Moreover, in 30% of cases the algorithm was not applicable or indeterminate. H2FPEF score had a modest correlation with PCWP (r=0.44, P<0.0001), and accuracy was 73%; NT‐proBNP correlated weakly with PCWP (r=0.24, P<0.001), and accuracy was 57%. Conclusions The 2016 ASE/EACVI algorithm for the assessment of diastolic function has a limited diagnostic accuracy in patients evaluated for unexplained dyspnea and/or pulmonary hypertension, and especially sensitivity to detect diastolic dysfunction was low.
Collapse
Affiliation(s)
- Arno A van de Bovenkamp
- Department of Cardiology Amsterdam Cardiovascular Sciences (ACS) Amsterdam UMC, Vrije Universiteit Amsterdam Amsterdam The Netherlands
| | - Vidya Enait
- Department of Cardiology Amsterdam Cardiovascular Sciences (ACS) Amsterdam UMC, Vrije Universiteit Amsterdam Amsterdam The Netherlands
| | - Frances S de Man
- Department of Pulmonology Amsterdam Cardiovascular Sciences (ACS) Amsterdam UMC, Vrije Universiteit Amsterdam Amsterdam The Netherlands
| | - Frank T P Oosterveer
- Department of Pulmonology Amsterdam Cardiovascular Sciences (ACS) Amsterdam UMC, Vrije Universiteit Amsterdam Amsterdam The Netherlands
| | - Harm Jan Bogaard
- Department of Pulmonology Amsterdam Cardiovascular Sciences (ACS) Amsterdam UMC, Vrije Universiteit Amsterdam Amsterdam The Netherlands
| | - Anton Vonk Noordegraaf
- Department of Pulmonology Amsterdam Cardiovascular Sciences (ACS) Amsterdam UMC, Vrije Universiteit Amsterdam Amsterdam The Netherlands
| | - Albert C van Rossum
- Department of Cardiology Amsterdam Cardiovascular Sciences (ACS) Amsterdam UMC, Vrije Universiteit Amsterdam Amsterdam The Netherlands
| | - M Louis Handoko
- Department of Cardiology Amsterdam Cardiovascular Sciences (ACS) Amsterdam UMC, Vrije Universiteit Amsterdam Amsterdam The Netherlands
| |
Collapse
|
40
|
Thomsen CF, Goharian TS, Larsen KT, Goetze JP, Andersen LB, Jeppesen JL. Intensive Lifestyle Intervention Increases Plasma Midregional Proatrial Natriuretic Peptide Concentrations in Overweight Children. J Am Heart Assoc 2021; 10:e020676. [PMID: 34180245 PMCID: PMC8403313 DOI: 10.1161/jaha.121.020676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Overweight adults have low circulating concentrations of ANP (atrial natriuretic peptide) and proANP fragments. We tested the hypothesis that an intensive lifestyle intervention with an intended weight loss would increase plasma concentrations of a proANP fragment in overweight children. Methods and Results We measured MR‐proANP (midregional proANP) concentrations in plasma from overweight children who participated in the OOIS (Odense Overweight Intervention Study). OOIS randomized 115 overweight children (11–13 years, 55% girls) to an intensive day‐camp intervention arm with increased physical activity and healthy diet or to a less intensive standard intervention arm for 6 weeks. We used linear mixed‐effects modeling for repeated measures to estimate the difference in the mean change with 95% CIs in fasting plasma MR‐proANP concentrations between the 2 arms, and we used partial least squares regression analysis to identify candidate mediators. Differences in weight, fitness, and metabolic factors were also analyzed. At baseline, fasting plasma MR‐proANP concentrations were (median [interquartile range]) 35.0 pmol/L (26.8–42.0) in the day‐camp intervention arm and 37.2 pmol/L (31.7–44.7) in standard intervention arm participants, respectively. After 6 weeks intervention, children in the day‐camp intervention arm had increased their MR‐proANP (5.4 pmol/L [0.8–10.0], P=0.022) and their fitness (2.33 mL O2/min per kg [0.52–4.14], P=0.012) and they had deceased their body mass index (−2.12 kg/m2 [−2.59 to −1.65], P<0.001) as compared with children in standard intervention arm. In the partial least squares analysis, decreases in fasting insulin and in estimated insulin resistance were associated with the observed increase in MR‐proANP concentrations. Conclusions An intensive lifestyle intervention increases plasma MR‐proANP among overweight children. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT01574352.
Collapse
Affiliation(s)
- Camilla F Thomsen
- Department of Medicine Amager Hvidovre Hospital in GlostrupUniversity of Copenhagen Glostrup Denmark
| | - Tina S Goharian
- Department of Clinical Biochemistry Nordsjællands Hospital HillerødUniversity of Copenhagen Hillerød Denmark
| | - Kristian T Larsen
- Center of Research in Childhood Health (RICH) Department of Sports Science and Clinical Biomechanics University of Southern Denmark Odense Denmark
| | - Jens P Goetze
- Department of Clinical Biochemistry Rigshospitalet BlegdamsvejUniversity of Copenhagen Denmark.,Department of Biomedical Sciences Faculty of Health and Medical Sciences University of Copenhagen Denmark
| | - Lars B Andersen
- Department of Sport, Food, and Natural Sciences Campus SogndalWestern Norway University of Applied Sciences, Sogndal Norway
| | - Jørgen L Jeppesen
- Department of Medicine Amager Hvidovre Hospital in GlostrupUniversity of Copenhagen Glostrup Denmark.,Department of Clinical Medicine Faculty of Health and Medical Sciences University of Copenhagen Denmark
| |
Collapse
|
41
|
Rukavina Mikusic NL, Kouyoumdzian NM, Puyó AM, Fernández BE, Choi MR. Role of natriuretic peptides in the cardiovascular-adipose communication: a tale of two organs. Pflugers Arch 2021; 474:5-19. [PMID: 34173888 DOI: 10.1007/s00424-021-02596-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 05/31/2021] [Accepted: 06/11/2021] [Indexed: 12/23/2022]
Abstract
Natriuretic peptides have long been known for their cardiovascular function. However, a growing body of evidence emphasizes the role of natriuretic peptides in the energy metabolism of several substrates in humans and animals, thus interrelating the heart, as an endocrine organ, with various insulin-sensitive tissues and organs such as adipose tissue, muscle skeletal, and liver. Adipose tissue dysfunction is associated with altered regulation of the natriuretic peptide system, also indicated as a natriuretic disability. Evidence points to a contribution of this natriuretic disability to the development of obesity, type 2 diabetes mellitus, and cardiometabolic complications; although the causal relationship is not fully understood at present. However, targeting the natriuretic peptide pathway may improve metabolic health in obesity and type 2 diabetes mellitus. This review will focus on the current literature on the metabolic functions of natriuretic peptides with emphasis on lipid metabolism and insulin sensitivity. Natriuretic peptide system alterations could be proposed as one of the linking mechanisms between adipose tissue dysfunction and cardiovascular disease.
Collapse
Affiliation(s)
- Natalia Lucía Rukavina Mikusic
- Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Nicolás Martín Kouyoumdzian
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana María Puyó
- Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Marcelo Roberto Choi
- Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto Universitario de Ciencias de la Salud, Fundación H.A. Barceló, Buenos Aires, Argentina
| |
Collapse
|
42
|
Galli E, Bourg C, Kosmala W, Oger E, Donal E. Phenomapping Heart Failure with Preserved Ejection Fraction Using Machine Learning Cluster Analysis: Prognostic and Therapeutic Implications. Heart Fail Clin 2021; 17:499-518. [PMID: 34051979 DOI: 10.1016/j.hfc.2021.02.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by a high rate of hospitalization and mortality (up to 84% at 5 years), which are similar to those observed for heart failure with reduced ejection fraction (HFrEF). These epidemiologic data claim for the development of specific and innovative therapies to reduce the burden of morbidity and mortality associated with this disease. Compared with HFrEF, which is due to a primary myocardial damage (eg ischemia, cardiomyopathies, toxicity), a heterogeneous etiologic background characterizes HFpEF. The authors discuss these phenotypes and specificities for defining therapeutic strategies that could be proposed according to phenotypes.
Collapse
Affiliation(s)
- Elena Galli
- University of Rennes, CHU Rennes, INSERM, LTSI-UMR 1099, Rennes F-35000, France
| | - Corentin Bourg
- University of Rennes, CHU Rennes, INSERM, LTSI-UMR 1099, Rennes F-35000, France
| | - Wojciech Kosmala
- Cardiology Department, Wroclaw Medical University, Wroclaw, Poland
| | - Emmanuel Oger
- University of Rennes, EA 7449 REPERES [Pharmacoepidemiology and Health Services Research], Rennes, France
| | - Erwan Donal
- University of Rennes, CHU Rennes, INSERM, LTSI-UMR 1099, Rennes F-35000, France.
| |
Collapse
|
43
|
Myhre PL, Lyngbakken MN, Berge T, Røysland R, Aagaard EN, Pervez O, Kvisvik B, Brynildsen J, Norseth J, Tveit A, Steine K, Omland T, Røsjø H. Diagnostic Thresholds for Pre-Diabetes Mellitus and Diabetes Mellitus and Subclinical Cardiac Disease in the General Population: Data From the ACE 1950 Study. J Am Heart Assoc 2021; 10:e020447. [PMID: 33998259 PMCID: PMC8483542 DOI: 10.1161/jaha.120.020447] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Diabetes mellitus (DM) is associated with left ventricular remodeling and incident heart failure, but the association between glycated hemoglobin A1c (HbA1c) and subclinical cardiac disease is not established. We aimed to determine the associations between HbA1c and (1) echocardiographic measures of left ventricular structure and function, and (2) cardiovascular biomarkers: cardiac troponin T, NT-proBNP (N-terminal pro-B-type natriuretic peptide), and CRP (C-reactive protein). Methods and Results Participants (n=3688) born in 1950 from the population-based ACE (Akershus Cardiac Examination) 1950 Study were classified as DM (HbA1c≥6.5% or self-reported DM), pre-DM (HbA1c 5.7%-6.5%), and no-DM (HbA1c<5.7%). DM, pre-DM, and no-DM were classified in 380 (10%), 1630 (44%), and 1678 (46%) participants, respectively. Mean age was 63.9±0.7 years, mean body mass index was 27.2±4.4 kg/m2, and 49% were women. Higher HbA1c was associated with worse left ventricular systolic (ejection fraction and global longitudinal strain) and diastolic (E/e'-ratio) function, myocardial injury (cardiac troponin T), inflammation (CRP), and impaired neurohormonal homeostasis (NT-proBNP) (P<0.001 in unadjusted and P<0.01 in adjusted analysis for all). The associations between HbA1c and cardiovascular biomarkers were independent of the echocardiographic variables, and vice versa. Associations were nonlinear (P<0.05 for nonlinearity) and appeared stronger in the pre-DM range of HbA1c than the no-DM and DM range. Conclusions HbA1c was associated with indexes of subclinical cardiovascular disease, and this was more pronounced in pre-DM. Our results suggest that cardiovascular preventive measures should be considered also in subjects with hyperglycemia and HbA1c below the established DM cutoff. Registration clinicaltrials.gov. Identifier: NCT01555411.
Collapse
Affiliation(s)
- Peder L Myhre
- Department of CardiologyAkershus University Hospital Lørenskog Norway.,Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway
| | - Magnus N Lyngbakken
- Department of CardiologyAkershus University Hospital Lørenskog Norway.,Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway
| | - Trygve Berge
- Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway.,Division for Research and InnovationAkershus University Hospital Lørenskog Norway
| | - Ragnhild Røysland
- Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway.,Institute for Clinical MedicineUniversity of Oslo Norway
| | - Erika N Aagaard
- Department of CardiologyAkershus University Hospital Lørenskog Norway.,Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway
| | - Osman Pervez
- Department of CardiologyAkershus University Hospital Lørenskog Norway.,Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway
| | - Brede Kvisvik
- Department of CardiologyAkershus University Hospital Lørenskog Norway.,Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway
| | - Jon Brynildsen
- Department of CardiologyAkershus University Hospital Lørenskog Norway.,Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway
| | - Jon Norseth
- Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway.,Department of Medical ResearchVestre Viken Hospital Trust Bærum Norway
| | - Arnljot Tveit
- Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway.,Division for Research and InnovationAkershus University Hospital Lørenskog Norway
| | - Kjetil Steine
- Department of CardiologyAkershus University Hospital Lørenskog Norway.,Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway
| | - Torbjørn Omland
- Department of CardiologyAkershus University Hospital Lørenskog Norway.,Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway
| | - Helge Røsjø
- Department of Multidisciplinary Laboratory Medicine and Medical BiochemistryAkershus University Hospital Lørenskog Norway.,Department of Laboratory MedicineVestre Viken Hospital Trust Bærum Norway
| |
Collapse
|
44
|
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
|
45
|
Ren J, Wu NN, Wang S, Sowers JR, Zhang Y. Obesity cardiomyopathy: evidence, mechanisms, and therapeutic implications. Physiol Rev 2021; 101:1745-1807. [PMID: 33949876 PMCID: PMC8422427 DOI: 10.1152/physrev.00030.2020] [Citation(s) in RCA: 190] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The prevalence of heart failure is on the rise and imposes a major health threat, in part, due to the rapidly increased prevalence of overweight and obesity. To this point, epidemiological, clinical, and experimental evidence supports the existence of a unique disease entity termed “obesity cardiomyopathy,” which develops independent of hypertension, coronary heart disease, and other heart diseases. Our contemporary review evaluates the evidence for this pathological condition, examines putative responsible mechanisms, and discusses therapeutic options for this disorder. Clinical findings have consolidated the presence of left ventricular dysfunction in obesity. Experimental investigations have uncovered pathophysiological changes in myocardial structure and function in genetically predisposed and diet-induced obesity. Indeed, contemporary evidence consolidates a wide array of cellular and molecular mechanisms underlying the etiology of obesity cardiomyopathy including adipose tissue dysfunction, systemic inflammation, metabolic disturbances (insulin resistance, abnormal glucose transport, spillover of free fatty acids, lipotoxicity, and amino acid derangement), altered intracellular especially mitochondrial Ca2+ homeostasis, oxidative stress, autophagy/mitophagy defect, myocardial fibrosis, dampened coronary flow reserve, coronary microvascular disease (microangiopathy), and endothelial impairment. Given the important role of obesity in the increased risk of heart failure, especially that with preserved systolic function and the recent rises in COVID-19-associated cardiovascular mortality, this review should provide compelling evidence for the presence of obesity cardiomyopathy, independent of various comorbid conditions, underlying mechanisms, and offer new insights into potential therapeutic approaches (pharmacological and lifestyle modification) for the clinical management of obesity cardiomyopathy.
Collapse
Affiliation(s)
- Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China.,Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Ne N Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| | - Shuyi Wang
- School of Medicine, Shanghai University, Shanghai, China.,University of Wyoming College of Health Sciences, Laramie, Wyoming
| | - James R Sowers
- Dalton Cardiovascular Research Center, Diabetes and Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
46
|
Pang Z, Pan C, Yao Z, Ren Y, Tian L, Cui J, Liu X, Zhang L, Chen Y. A study of the sequential treatment of acute heart failure with sacubitril/valsartan by recombinant human brain natriuretic peptide: A randomized controlled trial. Medicine (Baltimore) 2021; 100:e25621. [PMID: 33879733 PMCID: PMC8078236 DOI: 10.1097/md.0000000000025621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 04/03/2021] [Indexed: 01/04/2023] Open
Abstract
This study aimed to investigate the effects of the basic treatment for heart failure and sequential treatment with rh-brain natriuretic peptide (rhBNP) alone or the combination of rhBNP and sacubitril/valsartan. Cardiac structure, pulmonary artery pressure, inflammation and oxidative stress in patients with acute heart failure were evaluated.Three hundred patients with acute heart failure were included. According to the random number table method, the patients were divided into 3 groups of 100 patients per group: the standard treatment group (treated with an angiotensin-converting enzyme inhibitor, β receptor blocker, and corticosteroid antagonist), rhBNP group (basic treatment combined with rhBNP) and sequential treatment group (basic treatment for heart failure combined with rhBNP followed by sacubitril/valsartan). The changes in NT-probrain natriuretic peptide (BNP) levels, cardiac troponin T (cTnT) levels, cardiac structure, pulmonary artery pressure, and the levels inflammatory factors and oxidative stress factors were compared among the 3 groups at 1, 4, 12, and 36 weeks after treatment.The sequential treatment group displayed superior outcomes than the standard treatment group and the rhBNP group in terms of left atrium diameter, left ventricular end diastolic volume, left ventricular ejection fraction, pulmonary artery pressure, NT-proBNP levels, and cTnT levels, which respond to damage to the heart structure and myocardium. This result may be related to the decreased levels of inflammatory factors and the correction of oxidative stress imbalance.Sacubitril/valsartan significantly reduce the serum levels of inflammatory factors in patients with acute heart failure while decreasing the levels of oxidizing factors and increasing the levels of antioxidant factors. These changes may be one of the explanations for the better cardiac structure and better pulmonary artery pressure observed in the sequential treatment group.
Collapse
|
47
|
Aguilar M, Rose RA, Takawale A, Nattel S, Reilly S. New aspects of endocrine control of atrial fibrillation and possibilities for clinical translation. Cardiovasc Res 2021; 117:1645-1661. [PMID: 33723575 PMCID: PMC8208746 DOI: 10.1093/cvr/cvab080] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/25/2021] [Accepted: 03/11/2021] [Indexed: 12/20/2022] Open
Abstract
Hormones are potent endo-, para-, and autocrine endogenous regulators of the function of multiple organs, including the heart. Endocrine dysfunction promotes a number of cardiovascular diseases, including atrial fibrillation (AF). While the heart is a target for endocrine regulation, it is also an active endocrine organ itself, secreting a number of important bioactive hormones that convey significant endocrine effects, but also through para-/autocrine actions, actively participate in cardiac self-regulation. The hormones regulating heart-function work in concert to support myocardial performance. AF is a serious clinical problem associated with increased morbidity and mortality, mainly due to stroke and heart failure. Current therapies for AF remain inadequate. AF is characterized by altered atrial function and structure, including electrical and profibrotic remodelling in the atria and ventricles, which facilitates AF progression and hampers its treatment. Although features of this remodelling are well-established and its mechanisms are partly understood, important pathways pertinent to AF arrhythmogenesis are still unidentified. The discovery of these missing pathways has the potential to lead to therapeutic breakthroughs. Endocrine dysfunction is well-recognized to lead to AF. In this review, we discuss endocrine and cardiocrine signalling systems that directly, or as a consequence of an underlying cardiac pathology, contribute to AF pathogenesis. More specifically, we consider the roles of products from the hypothalamic-pituitary axis, the adrenal glands, adipose tissue, the renin–angiotensin system, atrial cardiomyocytes, and the thyroid gland in controlling atrial electrical and structural properties. The influence of endocrine/paracrine dysfunction on AF risk and mechanisms is evaluated and discussed. We focus on the most recent findings and reflect on the potential of translating them into clinical application.
Collapse
Affiliation(s)
- Martin Aguilar
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, QC, Canada.,Department of Pharmacology and Physiology/Institute of Biomedical Engineering, Université de Montréal, Montréal, QC, Canada
| | - Robert A Rose
- Department of Cardiac Sciences, Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, Health Research Innovation Center, University of Calgary, AB, Canada
| | - Abhijit Takawale
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, QC, Canada.,Department of Pharmacology and Physiology/Institute of Biomedical Engineering, Université de Montréal, Montréal, QC, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Stanley Nattel
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.,Faculty of Medicine, Department of Pharmacology and Physiology, and Research Centre, Montreal Heart Institute and University of Montreal, Montreal, QC, Canada.,Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Germany.,IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
48
|
The influence of sex and body mass index on the association between soluble neprilysin and risk of heart failure hospitalizations. Sci Rep 2021; 11:5940. [PMID: 33723360 PMCID: PMC7960699 DOI: 10.1038/s41598-021-85490-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 03/02/2021] [Indexed: 12/11/2022] Open
Abstract
A higher neprilysin activity has been suggested in women. In this retrospective analysis, we evaluated the association of sex and body mass index (BMI) with soluble neprilysin (sNEP) and recurrent admissions among 1021 consecutive HF outpatients. The primary and secondary endpoints were the number of HF hospitalizations and all-cause mortality, respectively. The association between sNEP with either endpoint was evaluated across sex and BMI categories (≥ 25 kg/m2 vs. < 25 kg/m2). Bivariate count regression (Poisson) was used, and risk estimates were expressed as incidence rates ratio (IRR). During a median follow-up of 6.65 years (percentile 25%-percentile 75%:2.83–10.25), 702 (68.76%) patients died, and 406 (40%) had at least 1 HF hospitalization. Median values of sNEP and BMI were 0.64 ng/mL (0.39–1.22), and 26.9 kg/m2 (24.3–30.4), respectively. Left ventricle ejection fraction was < 40% in 78.9% of patients, and 28% were women. In multivariable analysis, sNEP (main effect) was positively associated with HF hospitalizations (p = 0.001) but not with mortality (p = 0.241). The predictive value of sNEP for HF hospitalizations varied non-linearly across sex and BMI categories (p-value for interaction = 0.003), with significant and positive effect only on women with BMI ≥ 25 kg/m2 (p = 0.039). For instance, compared to men, women with sNEP of 1.22 ng/mL (percentile 75%) showed a significantly increased risk (IRRs: 1.26; 95% CI: 1.05–1.53). The interaction analysis for mortality did not support a differential prognostic effect for sNEP (p = 0.072). In conclusion, higher sNEP levels in overweight women better predicted an increased risk of HF hospitalization.
Collapse
|
49
|
Henke C, Haufe S, Ziehl D, Bornstein SR, Schulz-Menger J, Heni M, Engeli S, Jordan J, Birkenfeld AL. Low-fat hypocaloric diet reduces neprilysin in overweight and obese human subjects. ESC Heart Fail 2021; 8:938-942. [PMID: 33638612 PMCID: PMC8006681 DOI: 10.1002/ehf2.13220] [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: 03/30/2020] [Revised: 10/04/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
AIMS Neprilysin (NEP), a zinc metallopeptidase, degrades a variety of bioactive peptides including natriuretic peptides terminating their biological action on arterial blood pressure and natriuresis. Pharmacological inhibition of NEP reduces mortality in patients with heart failure with reduced ejection fraction. Physiological interventions reducing NEP levels are unknown in humans. Because obesity leads to increased NEP levels and increases the risk for heart failure, we hypothesized that weight loss reduces NEP concentrations in plasma and tissue. METHODS AND RESULTS We randomized overweight to obese human subjects to a low-fat or low-carbohydrate hypocaloric 6 month weight loss intervention. Soluble NEP was determined in plasma, and NEP mRNA was analysed from subcutaneous adipose tissue before and after diet. Low-fat diet-induced weight loss reduced soluble NEP levels from 0.83 ± 0.18 to 0.72 ± 0.18 μg/L (P = 0.038), while subcutaneous adipose tissue NEP mRNA expression was reduced by both dietary interventions [21% (P = 0.0057) by low-fat diet and 16% (P = 0.048) by low-carbohydrate diet]. We also analysed the polymorphisms of the gene coding for NEP, rs9827586 and rs701109, known to be associated with plasma NEP levels. For both single-nucleotide polymorphisms, minor allele carriers (A/A) had higher baseline plasma NEP levels (rs9827586: β = 0.53 ± 0.23, P < 0.0001; rs701109: β = 0.43 ± 0.22, P = 0.0016), and minor allele carriers of rs9827586 responded to weight loss with a larger NEP reduction (rs9827586: P = 0.0048). CONCLUSIONS Our study identifies weight loss via a hypocaloric low-fat diet as the first physiological intervention in humans to reduce NEP in plasma and adipose tissue. Specific single-nucleotide polymorphisms further contribute to the decrease. Our findings may help to explain the beneficial effect of weight loss on cardiac function in patients with heart failure.
Collapse
Affiliation(s)
- Christine Henke
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Sven Haufe
- Institute of Sports Medicine, Hannover Medical School, Hanover, Germany
| | - Doreen Ziehl
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Diabetes, School of Life Course Science and Medicine, King's College London, London, UK
| | - Jeanette Schulz-Menger
- Experimental and Clinical Research Center (ECRC), a joint collaboration between Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Department of Cardiology and Nephrology, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | - Martin Heni
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Section of Internal Medicine IV, Department of Diabetology, Endocrinology and Nephrology, University Hospital Tübingen, Ottfriet-Müller-Str. 10, Tübingen, 72076, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
| | - Stefan Engeli
- Institute of Clinical Pharmacology, Hannover Medical School, Hannover, Germany
| | - Jens Jordan
- Institute of Clinical Pharmacology, Hannover Medical School, Hannover, Germany.,Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Andreas L Birkenfeld
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Diabetes, School of Life Course Science and Medicine, King's College London, London, UK.,Section of Internal Medicine IV, Department of Diabetology, Endocrinology and Nephrology, University Hospital Tübingen, Ottfriet-Müller-Str. 10, Tübingen, 72076, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
| |
Collapse
|
50
|
Chakraborty S, Ong WK, Yau WWY, Zhou Z, Bhanu Prakash KN, Toh SA, Han W, Yen PM, Sugii S. CD10 marks non-canonical PPARγ-independent adipocyte maturation and browning potential of adipose-derived stem cells. Stem Cell Res Ther 2021; 12:109. [PMID: 33541392 PMCID: PMC7863460 DOI: 10.1186/s13287-021-02179-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/20/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Effective stem cell therapy is dependent on the stem cell quality that is determined by their differentiation potential, impairment of which leads to poor engraftment and survival into the target cells. However, limitations in our understanding and the lack of reliable markers that can predict their maturation efficacies have hindered the development of stem cells as an effective therapeutic strategy. Our previous study identified CD10, a pro-adipogenic, depot-specific prospective cell surface marker of human adipose-derived stem cells (ASCs). Here, we aim to determine if CD10 can be used as a prospective marker to predict mature adipocyte quality and play a direct role in adipocyte maturation. METHODS We first generated 14 primary human subject-derived ASCs and stable immortalized CD10 knockdown and overexpression lines for 4 subjects by the lentiviral transduction system. To evaluate the role of CD10 in adipogenesis, the adipogenic potential of the human subject samples were scored against their respective CD10 transcript levels. Assessment of UCP1 expression levels was performed to correlate CD10 levels to the browning potential of mature ASCs. Quantitative polymerase chain reaction (qPCR) and Western blot analysis were performed to determine CD10-dependent regulation of various targets. Seahorse analysis of oxidative metabolism and lipolysis assay were studied. Lastly, as a proof-of-concept study, we used CD10 as a prospective marker for screening nuclear receptor ligands library. RESULTS We identified intrinsic CD10 levels as a positive determinant of adipocyte maturation as well as browning potential of ASCs. Interestingly, CD10 regulates ASC's adipogenic maturation non-canonically by modulating endogenous lipolysis without affecting the classical peroxisome proliferator-activated receptor gamma (PPARγ)-dependent adipogenic pathways. Furthermore, our CD10-mediated screening analysis identified dexamethasone and retinoic acid as stimulator and inhibitor of adipogenesis, respectively, indicating CD10 as a useful biomarker for pro-adipogenic drug screening. CONCLUSION Overall, we establish CD10 as a functionally relevant ASC biomarker, which may be a prerequisite to identify high-quality cell populations for improving metabolic diseases.
Collapse
Affiliation(s)
- Smarajit Chakraborty
- Fat Metabolism and Stem Cell Group, Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR) Singapore, 11 Biopolis Way, Singapore, 138667, Singapore
- Xenobiology Division, Institute of Bioengineering and Nanotechnology (IBN) Singapore, A*STAR, 31 Biopolis Way, Singapore, 138669, Singapore
| | - Wee Kiat Ong
- Fat Metabolism and Stem Cell Group, Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR) Singapore, 11 Biopolis Way, Singapore, 138667, Singapore
- School of Pharmacy, Monash University Malaysia, 47500, Selangor, Malaysia
| | - Winifred W Y Yau
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School Singapore, 8 College Road, Singapore, 169857, Singapore
| | - Zhihong Zhou
- Fat Metabolism and Stem Cell Group, Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR) Singapore, 11 Biopolis Way, Singapore, 138667, Singapore
| | - K N Bhanu Prakash
- Signal and Image Processing Group, SBIC, A*STAR Singapore, 11 Biopolis Way, Singapore, 138667, Singapore
| | - Sue-Anne Toh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 5 Lower Kent Ridge Road, Singapore, 119074, Singapore
| | - Weiping Han
- Laboratory of Metabolic Medicine, SBIC, A*STAR Singapore, 11 Biopolis Way, Singapore, 138667, Singapore
| | - Paul M Yen
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School Singapore, 8 College Road, Singapore, 169857, Singapore
| | - Shigeki Sugii
- Fat Metabolism and Stem Cell Group, Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR) Singapore, 11 Biopolis Way, Singapore, 138667, Singapore.
- Xenobiology Division, Institute of Bioengineering and Nanotechnology (IBN) Singapore, A*STAR, 31 Biopolis Way, Singapore, 138669, Singapore.
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School Singapore, 8 College Road, Singapore, 169857, Singapore.
| |
Collapse
|