101
|
Frank AP, Palmer BF, Clegg DJ. Do estrogens enhance activation of brown and beiging of adipose tissues? Physiol Behav 2017; 187:24-31. [PMID: 28988965 DOI: 10.1016/j.physbeh.2017.09.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/30/2017] [Accepted: 09/30/2017] [Indexed: 01/05/2023]
Abstract
Obesity and its associated co-morbidities are worldwide public health concerns. Obesity is characterized by excessive adipose tissue accumulation; however, it is important to recognize that human and rodent adipose tissues are made up of several distinct adipose tissue sub-types. White adipose tissue (WAT) is considered the prototypical fat cell, due to its capacity and capability to store large amounts of lipid. In contrast, brown adipose tissue (BAT) oxidizes substrates to generate heat. BAT contains more mitochondria than WAT and express uncoupling protein-1 (UCP1), which mediates BAT thermogenesis. A third sub-type of adipose tissue, Brown-in-white (BRITE)/beige adipocytes arise from WAT upon adrenergic stimulation and resembles BAT functionally. The energy burning feature of BAT/beige cells, combined with evidence of an inverse-correlation between BAT/beige adipose tissue and obesity have given rise to the hypothesis that obesity may be linked to BAT/beige 'malfunction'. Females have more BAT and perhaps an enhanced capacity to beige their adipose tissue when compared to males. Multiple signal pathways are capable of activating BAT thermogenesis and beiging of WAT; here, we discuss the potential role of estrogens in enhancing and mediating these factors to enhance adipose tissue thermogenesis.
Collapse
Affiliation(s)
- Aaron P Frank
- Biomedical Research Division, Diabetes and Obesity Research Institute, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Biff F Palmer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Deborah J Clegg
- Biomedical Research Division, Diabetes and Obesity Research Institute, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
102
|
Baldassarre S, Fragapani S, Panero A, Fedele D, Pinach S, Lucchiari M, Vitale AR, Mengozzi G, Gruden G, Bruno G. NTproBNP in insulin-resistance mediated conditions: overweight/obesity, metabolic syndrome and diabetes. The population-based Casale Monferrato Study. Cardiovasc Diabetol 2017; 16:119. [PMID: 28946871 PMCID: PMC5613356 DOI: 10.1186/s12933-017-0601-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/17/2017] [Indexed: 01/03/2023] Open
Abstract
Background and aims NTproBNP and BNP levels are reduced in obese subjects, but population-based data comparing the pattern of this relationship in the full spectrum of insulin-resistance mediated conditions, overweight/obesity, metabolic syndrome and diabetes, are limited. Methods The study-base were 3244 individuals aged 45–74 years, none of whom had heart failure, 1880 without diabetes and 1364 with diabetes, identified as part of two surveys of the population-based Casale Monferrato Study. All measurements were centralized. We examined with multiple linear regression and cubic regression splines the relationship between NTproBNP and BMI, independently of known risk factors and confounders. A logistic regression analysis was also performed to assess the effect of overweight/obesity (BMI ≥ 25 kg/m2), diabetes and metabolic syndrome on NTproBNP values. Results Out of the overall cohort of 3244 people, overweight/obesity was observed in 1118 (59.4%) non-diabetic and 917 (67.2%) diabetic subjects, respectively. In logistic regression, compared to normal weight individuals, those with a BMI ≥ 25 kg/m2 had a OR of 0.70 (95% CI 0.56–0.87) of having high NTproBNP values, independently of diabetes. As interaction between diabetes and NTproBNP was evident (p < 0.001), stratified analyses were performed. Diabetes either alone or combined with overweight/obesity or metabolic syndrome enhanced fourfold and over the OR of having high NTproBNP levels, while the presence of metabolic syndrome alone had a more modest effect (OR 1.54, 1.18–2.01) even after having excluded individuals with CVD. In the non-diabetic cohort, obesity/overweight and HOMA-IR ≥ 2.0 decreased to a similar extent the ORs of high NTproBNP [0.76 (0.60–0.95) and 0.74 (0.59–0.93)], but the association between overweight/obesity and NTproBNP was no longer significant after the inclusion into the model of HOMA-IR, whereas CRP > 3 mg/dl conferred a fully adjusted OR of 0.65 (0.49–0.86). Conclusions NT-proBNP levels are lower in overweight/obesity, even in those with diabetes. Both insulin-resistance and chronic low-grade inflammation are involved in this relationship. Further intervention studies are required to clarify the potential role of drugs affecting the natriuretic peptides system on body weight and risk of diabetes.
Collapse
Affiliation(s)
- Stefano Baldassarre
- Dept. of Medical Sciences, University of Torino, corso Dogliotti 14, 10126, Turin, Italy
| | - Salvatore Fragapani
- Dept. of Medical Sciences, University of Torino, corso Dogliotti 14, 10126, Turin, Italy
| | - Antonio Panero
- Dept. of Medical Sciences, University of Torino, corso Dogliotti 14, 10126, Turin, Italy
| | - Debora Fedele
- Dept. of Medical Sciences, University of Torino, corso Dogliotti 14, 10126, Turin, Italy
| | - Silvia Pinach
- Dept. of Medical Sciences, University of Torino, corso Dogliotti 14, 10126, Turin, Italy
| | - Manuela Lucchiari
- Clinical Chemistry Laboratory, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Anna Rita Vitale
- Clinical Chemistry Laboratory, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Giulio Mengozzi
- Clinical Chemistry Laboratory, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Gabriella Gruden
- Dept. of Medical Sciences, University of Torino, corso Dogliotti 14, 10126, Turin, Italy
| | - Graziella Bruno
- Dept. of Medical Sciences, University of Torino, corso Dogliotti 14, 10126, Turin, Italy.
| |
Collapse
|
103
|
Makihara H, Hidaka M, Sakai Y, Horie Y, Mitsui H, Ohashi K, Goshima Y, Akase T. Reduction and fragmentation of elastic fibers in the skin of obese mice is associated with altered mRNA expression levels of fibrillin-1 and neprilysin. Connect Tissue Res 2017; 58:479-486. [PMID: 27892729 DOI: 10.1080/03008207.2016.1255205] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
AIM OF THE STUDY Our previous research suggested that obesity induces structural fragility in the skin. Elastic fibers impart strength and elasticity. In this study, we determined whether elastic fibers decrease in the skin of obese mice. MATERIALS AND METHODS To confirm alterations in elastic fiber content due to obesity, we used spontaneously obese model mice (TSOD) and control mice (TSNO). Furthermore, to evaluate the elastin structure and gene expression dependent on the severity of obesity, an obesity-enhanced mouse model was developed by feeding a high fat diet to TSOD (TSOD-HF). Back skin samples were stained with hematoxylin and eosin and Elastica van Gieson for microscopic examination, and the samples were stained for immunohistochemical analysis of neprilysin. Gene expression levels were determined using a real-time PCR system. RESULTS The abundance of elastic fibers beneath the epidermis was remarkably reduced and fragmented in TSOD as compared with TSNO. Fibrillin-1 mRNA levels in TSOD were significantly suppressed compared with those in TSNO, whereas neprilysin mRNA levels and immunohistochemical expression in TSOD were significantly increased, as compared with those in TSNO. The reduction of elastic fibers was enhanced and the expression levels of elastic fiber formed factors were significantly suppressed in TSOD-HF, as compared with those in the TSOD. CONCLUSIONS The abundance of elastic fibers was reduced and fragmented in obesity, suggesting that the reduction in elastic fibers is initially caused by increased neprilysin and decreased fibrillin-1 expression, which may inhibit formation and stabilization of elastic fibers, resulting in skin fragility in obesity.
Collapse
Affiliation(s)
- Hiroko Makihara
- a Department of Molecular Pharmacology and Neurobiology , Yokohama City University Graduate School of Medicine , Yokohama , Japan.,b Department of Biological Science and Nursing , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Moeko Hidaka
- b Department of Biological Science and Nursing , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Yui Sakai
- b Department of Biological Science and Nursing , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Yoshiko Horie
- b Department of Biological Science and Nursing , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Hideaki Mitsui
- c Department of Pathology , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Kenichi Ohashi
- c Department of Pathology , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Yoshio Goshima
- a Department of Molecular Pharmacology and Neurobiology , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Tomoko Akase
- b Department of Biological Science and Nursing , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| |
Collapse
|
104
|
Urinary sodium excretion after gastric bypass surgery. Surg Obes Relat Dis 2017; 13:1506-1514. [DOI: 10.1016/j.soard.2017.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/09/2017] [Accepted: 04/03/2017] [Indexed: 12/28/2022]
|
105
|
GLP-1 receptor independent pathways: emerging beneficial effects of GLP-1 breakdown products. Eat Weight Disord 2017; 22:231-240. [PMID: 28040864 DOI: 10.1007/s40519-016-0352-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 12/15/2016] [Indexed: 12/26/2022] Open
Abstract
The glucagon-like peptide-1 (GLP-1) axis has emerged as a major therapeutic target for the treatment of type 2 diabetes and, recently, of obesity. The insulinotropic activity of the native incretin hormone GLP-1(7-36)amide, which is mainly exerted through a unique G protein-coupled receptor (GLP-1R), is terminated via enzymatic cleavage by dipeptidyl peptidase-IV that generates a C-terminal GLP-1 metabolite GLP-1(9-36)amide, the major circulating form in plasma. GLP-1(28-36)amide and GLP-1(32-36)amide are further cleavage products derived from GLP-1(7-36)amide and GLP-1(9-36)amide by the action of a neutral endopeptidase known as neprilysin. Until recently, GLP-1-derived metabolites were generally considered metabolically inactive. However, emerging evidence indicates that GLP-1 byproducts have insulinomimetic activities that may contribute to the pleiotropic effects of GLP-1 independently of the canonical GLP-1R. The recent studies reporting the beneficial effects of the administration of these metabolites in vivo and in vitro are the focus of this review. Collectively, these results suggest that GLP-1 metabolites inhibit hepatic glucose production, exert antioxidant cardio- and neuroprotective actions, reduce oxidative stress in vasculature and have both anti-apoptotic and proliferative effects in pancreatic β-cells, putatively by the modulation of mitochondrial functions. These findings have implication in energy homeostasis, obesity and its associated metabolic and cardiovascular complications as well as incretin-based therapies for the treatment of diabetes and obesity.
Collapse
|
106
|
Moser VA, Pike CJ. Obesity Accelerates Alzheimer-Related Pathology in APOE4 but not APOE3 Mice. eNeuro 2017; 4:ENEURO.0077-17.2017. [PMID: 28612048 PMCID: PMC5469027 DOI: 10.1523/eneuro.0077-17.2017] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/17/2017] [Accepted: 05/22/2017] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD) risk is modified by both genetic and environmental risk factors, which are believed to interact to cooperatively modify pathogenesis. Although numerous genetic and environmental risk factors for AD have been identified, relatively little is known about potential gene-environment interactions in regulating disease risk. The strongest genetic risk factor for late-onset AD is the ε4 allele of apolipoprotein E (APOE4). An important modifiable risk factor for AD is obesity, which has been shown to increase AD risk in humans and accelerate development of AD-related pathology in rodent models. Potential interactions between APOE4 and obesity are suggested by the literature but have not been thoroughly investigated. In the current study, we evaluated this relationship by studying the effects of diet-induced obesity (DIO) in the EFAD mouse model, which combines familial AD transgenes with human APOE3 or APOE4. Male E3FAD and E4FAD mice were maintained for 12 weeks on either a control diet or a Western diet high in saturated fat and sugars. We observed that metabolic outcomes of DIO were similar in E3FAD and E4FAD mice. Importantly, our data showed a significant interaction between diet and APOE genotype on AD-related outcomes in which Western diet was associated with robust increases in amyloid deposits, β-amyloid burden, and glial activation in E4FAD but not in E3FAD mice. These findings demonstrate an important gene-environment interaction in an AD mouse model that suggests that AD risk associated with obesity is strongly influenced by APOE genotype.
Collapse
Affiliation(s)
- V Alexandra Moser
- Neuroscience Graduate Program, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| | - Christian J Pike
- Neuroscience Graduate Program, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
107
|
Willard JR, Barrow BM, Zraika S. Improved glycaemia in high-fat-fed neprilysin-deficient mice is associated with reduced DPP-4 activity and increased active GLP-1 levels. Diabetologia 2017; 60:701-708. [PMID: 27933334 PMCID: PMC5342915 DOI: 10.1007/s00125-016-4172-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 11/16/2016] [Indexed: 12/11/2022]
Abstract
AIM/HYPOTHESIS Neprilysin, a widely expressed peptidase, is upregulated in metabolically altered states such as obesity and type 2 diabetes. Like dipeptidyl peptidase-4 (DPP-4), neprilysin can degrade and inactivate the insulinotropic peptide glucagon-like peptide-1 (GLP-1). Thus, we investigated whether neprilysin deficiency enhances active GLP-1 levels and improves glycaemia in a mouse model of high fat feeding. METHODS Nep +/+ and Nep -/- mice were fed a 60% fat diet for 16 weeks, after which active GLP-1 and DPP-4 activity levels were measured, as were glucose, insulin and C-peptide levels during an OGTT. Insulin sensitivity was assessed using an insulin tolerance test. RESULTS High-fat-fed Nep -/- mice exhibited elevated active GLP-1 levels (5.8 ± 1.1 vs 3.5 ± 0.8 pmol/l, p < 0.05) in association with improved glucose tolerance, insulin sensitivity and beta cell function compared with high-fat-fed Nep +/+ mice. In addition, plasma DPP-4 activity was lower in high-fat-fed Nep -/- mice (7.4 ± 1.0 vs 10.7 ± 1.3 nmol ml-1 min-1, p < 0.05). No difference in insulin:C-peptide ratio was observed between Nep -/- and Nep +/+ mice, suggesting that improved glycaemia does not result from changes in insulin clearance. CONCLUSIONS/INTERPRETATION Under conditions of increased dietary fat, an improved glycaemic status in neprilysin-deficient mice is associated with elevated active GLP-1 levels, reduced plasma DPP-4 activity and improved beta cell function. Thus, neprilysin inhibition may be a novel treatment strategy for type 2 diabetes.
Collapse
Affiliation(s)
- Joshua R Willard
- Veterans Affairs Puget Sound Health Care System, 1660 South Columbian Way (151), Seattle, WA, 98108, USA
| | - Breanne M Barrow
- Veterans Affairs Puget Sound Health Care System, 1660 South Columbian Way (151), Seattle, WA, 98108, USA
| | - Sakeneh Zraika
- Veterans Affairs Puget Sound Health Care System, 1660 South Columbian Way (151), Seattle, WA, 98108, USA.
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
108
|
Campos-Peña V, Toral-Rios D, Becerril-Pérez F, Sánchez-Torres C, Delgado-Namorado Y, Torres-Ossorio E, Franco-Bocanegra D, Carvajal K. Metabolic Syndrome as a Risk Factor for Alzheimer's Disease: Is Aβ a Crucial Factor in Both Pathologies? Antioxid Redox Signal 2017; 26:542-560. [PMID: 27368351 DOI: 10.1089/ars.2016.6768] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Recently, chronic degenerative diseases have become one of the main health problems worldwide. That is the case of Alzheimer's disease (AD) and metabolic syndrome (MetS), whose expression can be influenced by different risk factors. Recent Advances: In recent decades, it has been widely described that MetS increases the risk of cognitive impairment and dementia. MetS pathogenesis involves several vascular risk factors such as diabetes, dyslipidemia, hypertension, and insulin resistance (I/R). CRITICAL ISSUES Reported evidence shows that vascular risk factors are associated with AD, particularly in the development of protein aggregation, inflammation, oxidative stress, neuronal dysfunction, and disturbances in signaling pathways, with insulin receptor signaling being a common alteration between MetS and AD. FUTURE DIRECTIONS Insulin signaling has been involved in tau phosphorylation and amyloid β (Aβ) metabolism. However, it has also been demonstrated that Aβ oligomers can bind to insulin receptors, triggering their internalization, decreasing neuron responsiveness to insulin, and promoting insulin I/R. Thus, it could be argued that Aβ could be a convergent factor in the development of both pathologies. Antioxid. Redox Signal. 26, 542-560.
Collapse
Affiliation(s)
| | - Danira Toral-Rios
- 2 Departamento de Fisiología Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Mexico City, Mexico
| | | | - Carmen Sánchez-Torres
- 4 Departamento of Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Mexico City, Mexico
| | | | - Elimar Torres-Ossorio
- 6 Facultad de Química, Universidad Nacional Autónoma de México , Mexico City, Mexico
| | | | - Karla Carvajal
- 7 Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría , Mexico City, Mexico
| |
Collapse
|
109
|
Neprilysin facilitates adipogenesis through potentiation of the phosphatidylinositol 3-kinase (PI3K) signaling pathway. Mol Cell Biochem 2017; 430:1-9. [DOI: 10.1007/s11010-017-2948-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/17/2017] [Indexed: 12/28/2022]
|
110
|
Jordan J, Stinkens R, Jax T, Engeli S, Blaak EE, May M, Havekes B, Schindler C, Albrecht D, Pal P, Heise T, Goossens GH, Langenickel TH. Improved Insulin Sensitivity With Angiotensin Receptor Neprilysin Inhibition in Individuals With Obesity and Hypertension. Clin Pharmacol Ther 2016; 101:254-263. [DOI: 10.1002/cpt.455] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/27/2016] [Accepted: 08/12/2016] [Indexed: 12/16/2022]
Affiliation(s)
- J Jordan
- Institute for Clinical Pharmacology, Hannover Medical School; Hannover Germany
| | - R Stinkens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism; Maastricht University Medical Center; Maastricht The Netherlands
| | | | - S Engeli
- Institute for Clinical Pharmacology, Hannover Medical School; Hannover Germany
| | - EE Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism; Maastricht University Medical Center; Maastricht The Netherlands
| | - M May
- Institute for Clinical Pharmacology, Hannover Medical School; Hannover Germany
| | - B Havekes
- Department of Internal Medicine, Division of Endocrinology; Maastricht University Medical Center; Maastricht The Netherlands
| | - C Schindler
- Institute for Clinical Pharmacology, Hannover Medical School; Hannover Germany
| | - D Albrecht
- Translational Medicine, Novartis Pharma AG; Basel Switzerland
| | - P Pal
- Biostatistical Sciences, Integrated Development Functions and Regions; Novartis Healthcare Pvt. Ltd; Hyderabad India
| | | | - GH Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism; Maastricht University Medical Center; Maastricht The Netherlands
| | - TH Langenickel
- Translational Medicine, Novartis Pharma AG; Basel Switzerland
| |
Collapse
|
111
|
Brain-Wide Insulin Resistance, Tau Phosphorylation Changes, and Hippocampal Neprilysin and Amyloid-β Alterations in a Monkey Model of Type 1 Diabetes. J Neurosci 2016; 36:4248-58. [PMID: 27076423 DOI: 10.1523/jneurosci.4640-14.2016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/02/2016] [Indexed: 02/08/2023] Open
Abstract
UNLABELLED Epidemiological findings suggest that diabetic individuals are at a greater risk for developing Alzheimer's disease (AD). To examine the mechanisms by which diabetes mellitus (DM) may contribute to AD pathology in humans, we examined brain tissue from streptozotocin-treated type 1 diabetic adult male vervet monkeys receiving twice-daily exogenous insulin injections for 8-20 weeks. We found greater inhibitory phosphorylation of insulin receptor substrate 1 in each brain region examined of the diabetic monkeys when compared with controls, consistent with a pattern of brain insulin resistance that is similar to that reported in the human AD brain. Additionally, a widespread increase in phosphorylated tau was seen, including brain areas vulnerable in AD, as well as relatively spared structures, such as the cerebellum. An increase in active ERK1/2 was also detected, consistent with DM leading to changes in tau-kinase activity broadly within the brain. In contrast to these widespread changes, we found an increase in soluble amyloid-β (Aβ) levels that was restricted to the temporal lobe, with the greatest increase seen in the hippocampus. Consistent with this localized Aβ increase, a hippocampus-restricted decrease in the protein and mRNA for the Aβ-degrading enzyme neprilysin (NEP) was found, whereas various Aβ-clearing and -degrading proteins were unchanged. Thus, we document multiple biochemical changes in the insulin-controlled DM monkey brain that can link DM with the risk of developing AD, including dysregulation of the insulin-signaling pathway, changes in tau phosphorylation, and a decrease in NEP expression in the hippocampus that is coupled with a localized increase in Aβ. SIGNIFICANCE STATEMENT Given that diabetes mellitus (DM) appears to increase the risk of developing Alzheimer's disease (AD), understanding the mechanisms by which DM promotes AD is important. We report that DM in a nonhuman primate brain leads to changes in the levels or posttranslational processing of proteins central to AD pathobiology, including tau, amyloid-β (Aβ), and the Aβ-degrading protease neprilysin. Additional evidence from this model suggests that alterations in brain insulin signaling occurred that are reminiscent of insulin signaling pathway changes seen in human AD. Thus, in an in vivo model highly relevant to humans, we show multiple alterations in the brain resulting from DM that are mechanistically linked to AD risk.
Collapse
|
112
|
Abstract
cGMP controls many cellular functions ranging from growth, viability, and differentiation to contractility, secretion, and ion transport. The mammalian genome encodes seven transmembrane guanylyl cyclases (GCs), GC-A to GC-G, which mainly modulate submembrane cGMP microdomains. These GCs share a unique topology comprising an extracellular domain, a short transmembrane region, and an intracellular COOH-terminal catalytic (cGMP synthesizing) region. GC-A mediates the endocrine effects of atrial and B-type natriuretic peptides regulating arterial blood pressure/volume and energy balance. GC-B is activated by C-type natriuretic peptide, stimulating endochondral ossification in autocrine way. GC-C mediates the paracrine effects of guanylins on intestinal ion transport and epithelial turnover. GC-E and GC-F are expressed in photoreceptor cells of the retina, and their activation by intracellular Ca(2+)-regulated proteins is essential for vision. Finally, in the rodent system two olfactorial GCs, GC-D and GC-G, are activated by low concentrations of CO2and by peptidergic (guanylins) and nonpeptidergic odorants as well as by coolness, which has implications for social behaviors. In the past years advances in human and mouse genetics as well as the development of sensitive biosensors monitoring the spatiotemporal dynamics of cGMP in living cells have provided novel relevant information about this receptor family. This increased our understanding of the mechanisms of signal transduction, regulation, and (dys)function of the membrane GCs, clarified their relevance for genetic and acquired diseases and, importantly, has revealed novel targets for therapies. The present review aims to illustrate these different features of membrane GCs and the main open questions in this field.
Collapse
Affiliation(s)
- Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
113
|
O'Neill S, Bohl M, Gregersen S, Hermansen K, O'Driscoll L. Blood-Based Biomarkers for Metabolic Syndrome. Trends Endocrinol Metab 2016; 27:363-374. [PMID: 27150849 DOI: 10.1016/j.tem.2016.03.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 03/10/2016] [Accepted: 03/20/2016] [Indexed: 01/08/2023]
Abstract
Metabolic syndrome (MetS) is a constellation of factors increasing the risk of type 2 diabetes mellitus (T2DM), cardiovascular disease (CVD), and cancer. MetS diagnosis is cumbersome and the precise diagnosis differs throughout the world. Efforts are underway to find MetS biomarkers that could all be analysed in a single blood sample. Here we review recent advances, including progress on circulating exosomes and microvesicles and their molecular contents, as well as DNA, RNAs, and proteins taken directly from blood samples. While additional research is now warranted to advance upon these findings, there is reason for optimising that such blood-based entities will be beneficial for MetS diagnosis and will help reduce risk of T2DM, CVD, and cancers, contributing both societal and economic benefit.
Collapse
Affiliation(s)
- Sadhbh O'Neill
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Mette Bohl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Soren Gregersen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Kjeld Hermansen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
114
|
Brachs M, Wiegand S, Leupelt V, Ernert A, Kintscher U, Jumpertz von Schwarzenberg R, Decker AM, Bobbert T, Hübner N, Chen W, Krude H, Spranger J, Mai K. ANP system activity predicts variability of fat mass reduction and insulin sensitivity during weight loss. Metabolism 2016; 65:935-43. [PMID: 27173472 DOI: 10.1016/j.metabol.2016.03.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 03/22/2016] [Accepted: 03/31/2016] [Indexed: 02/08/2023]
Abstract
INTRODUCTION In weight loss trials, a considerable inter-individual variability in reduction of fat mass and changes of insulin resistance is observed, even under standardized study conditions. The underlying mechanisms are not well understood. Given the metabolic properties of the atrial natriuretic peptide (ANP) system, we hypothesized that ANP signaling might be involved in this phenomenon by changes of ANP secretion or receptor balance. Therefore, we investigated the impact of systemic, adipose and myocellular ANP system on metabolic and anthropometric improvements during weight loss. METHODS We comprehensively investigated 143 subjects (31 male, 112 female) before and after a 3 month-standardized weight loss program. The time course of BMI, fat mass, insulin sensitivity, circulating mid-regional proANP (MR-proANP) levels as well as adipose and myocellular natriuretic receptor A (NPR-A) and C (NPR-C) mRNA expression were investigated. RESULTS BMI decreased by -12.6±3.7%. This was accompanied by a remarkable decrease of adipose NPR-C expression (1005.0±488.4 vs. 556.7±465.6; p<0.001) as well as a tendency towards increased adipose NPR-A expression (4644.7±946.8 vs. 4877.6±869.8; p=0.051). Weight loss induced changes in NPR-C (ΔNPR-C) was linked to relative reduction of total fat mass (ΔFM) (r=0.281; p<0.05), reduction of BMI (r=0.277; p<0.01), and increase of free fatty acids (ΔFFA) (r=-0.258; p<0.05). Basal NPR-C expression and weight loss induced ΔNPR-C independently explained 22.7% of ΔFM. In addition, ΔMR-proANP was independently associated with improvement of insulin sensitivity (standardized ß=0.246, p<0.01). DISCUSSION ANP receptor expression predicted the degree of weight loss induced fat mass reduction. Our comprehensive human data support that peripheral ANP signalling is involved in control of adipose tissue plasticity and function during weight loss. (Funded by the Deutsche Forschungsgemeinschaft (KFO281/2), the Berlin Institute of Health (BIH) and the German Centre for Cardiovascular Research (DZHK/BMBF); ClinicalTrials.gov number: NCT00850629).
Collapse
Affiliation(s)
- Maria Brachs
- Department of Endocrinology & Metabolism, Charite - Universitätsmedizin, Berlin, Germany; Charité-Center for Cardiovascular Research (CCR), Berlin, Germany; Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Susanna Wiegand
- Department of Pediatric Endocrinology and Diabetology, Charité Universitätsmedizin, Berlin, Germany
| | - Verena Leupelt
- Department of Endocrinology & Metabolism, Charite - Universitätsmedizin, Berlin, Germany; Charité-Center for Cardiovascular Research (CCR), Berlin, Germany
| | - Andrea Ernert
- Department of Pediatric Endocrinology and Diabetology, Charité Universitätsmedizin, Berlin, Germany
| | - Ulrich Kintscher
- Charité-Center for Cardiovascular Research (CCR), Berlin, Germany
| | - Reiner Jumpertz von Schwarzenberg
- Department of Endocrinology & Metabolism, Charite - Universitätsmedizin, Berlin, Germany; Charité-Center for Cardiovascular Research (CCR), Berlin, Germany; Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Anne-Marie Decker
- Department of Endocrinology & Metabolism, Charite - Universitätsmedizin, Berlin, Germany; Charité-Center for Cardiovascular Research (CCR), Berlin, Germany; Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Thomas Bobbert
- Department of Endocrinology & Metabolism, Charite - Universitätsmedizin, Berlin, Germany; Charité-Center for Cardiovascular Research (CCR), Berlin, Germany
| | - Norbert Hübner
- Max Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| | - Wei Chen
- Max Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| | - Heiko Krude
- Department of Pediatric Endocrinology and Diabetology, Charité Universitätsmedizin, Berlin, Germany
| | - Joachim Spranger
- Department of Endocrinology & Metabolism, Charite - Universitätsmedizin, Berlin, Germany; Charité-Center for Cardiovascular Research (CCR), Berlin, Germany; Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Knut Mai
- Department of Endocrinology & Metabolism, Charite - Universitätsmedizin, Berlin, Germany; Charité-Center for Cardiovascular Research (CCR), Berlin, Germany; Experimental and Clinical Research Center (ECRC), Berlin, Germany.
| |
Collapse
|
115
|
Natriuretic peptide control of energy balance and glucose homeostasis. Biochimie 2016; 124:84-91. [DOI: 10.1016/j.biochi.2015.05.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/19/2015] [Indexed: 12/27/2022]
|
116
|
Katsanos S, Bistola V, Parissis JT. Combining angiotensin II receptor 1 antagonism and neprilysin inhibition for the treatment of heart failure. Expert Rev Clin Pharmacol 2016; 9:513-523. [PMID: 26873036 DOI: 10.1586/17512433.2016.1153423] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sacubitril/valsartan is a novel, first-in-class drug, which combines a neprilysin inhibitor with an angiotensin receptor blocker. Sacubitril inhibits neprilysin endopeptidase, blocking the catabolism of natriuretic peptides (NP), thereby increasing their bioavailability. Valsartan counterbalances the increase of angiotensin II that results from neprilysin inhibition, exerting also the beneficial effects of angiotensin receptor blockers seen in previous HF trials. PARADIGM-HF trial has proved the superiority of sacubitril/valsartan (LCZ696) over ACE inhibitor enalapril to reduce mortality and morbidity of symptomatic HF patients with reduced ejection fraction (HFrEF), setting the grounds for the replacement of ACE inhibitors by sacubitril/valsartan in the management of HFrEF. Sacubitril/valsartan is currently being tested in a phase III trial (PARAGON-HF) in patients with HF with preserved EF. PARAGON-HF is also expected to provide further data regarding the long-term safety of sacubitril/valsartan, hopefully to alleviate concerns regarding the effects of neprilysin inhibition on cognitive function.
Collapse
Affiliation(s)
- Spyridon Katsanos
- a Heart Failure Unit , Attikon University Hospital , Athens , Greece
| | - Vasiliki Bistola
- a Heart Failure Unit , Attikon University Hospital , Athens , Greece
| | - John T Parissis
- a Heart Failure Unit , Attikon University Hospital , Athens , Greece
| |
Collapse
|
117
|
Ul-Haq Z, Usmani S, Iqbal S, Zia SR. In silico based investigation of dynamic variations in neprilysin (NEP and NEP2) proteins for extracting the point of specificity. MOLECULAR BIOSYSTEMS 2016; 12:1024-36. [PMID: 26846903 DOI: 10.1039/c5mb00727e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neprilysin-2 (NEP2) in the central nervous system controls Alzheimer's protein (amyloid-β) deposition, and prevents its occurrence. However, in the peripheral system, its closest homolog, neutral endopeptidase (NEP), regulates hypertension and heart related diseases. Inhibitors of NEP with a lesser degree of specificity can treat hypertension with an increased risk of cerebral deposition of amyloid-β. In order to rationalize the point of selectivity, the dynamic behavior of human NEP and NEP2 proteins was monitored by conducting molecular dynamics (MD) simulations. A computationally reliable model of NEP2 was achieved with 79.9%, 19.1% and 0.2% residues in the allowed, additionally allowed and disallowed regions respectively, using as a reference protein. Additionally, molecular docking studies were carried out for a set of five already known inhibitors of NEP and modeled NEP2 to obtain the comparative behaviors of the complexes. MD results highlighted their different responses along with important residues having a part in ligand-protein binding. For substrate and inhibitor binding, Arg664/661 and Zn697/694 were identified as the most conserved residues. High degree flexible transitions during the MD simulations were also observed in loop areas along with active site residues. Energy calculations, hydrogen bonds and their occupancy rates helped to conclude each ligand's potency towards a particular target. In most complexes of hNEP2, the ligands showed weak interactions which might be due to its larger pocket size or huge conformational variations in active site residues upon complexation. In the case of inhibitors of a small size like thiorphan, Arg49 and Arg664 are found to be acting to support the ligand binding in NEP while only Arg661 is acting in NEP2.
Collapse
Affiliation(s)
- Zaheer Ul-Haq
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan.
| | - Saman Usmani
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan.
| | - Sadaf Iqbal
- Department of Chemistry, University of Karachi, Karachi-75270, Pakistan
| | - Syeda Rehana Zia
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan.
| |
Collapse
|
118
|
Abstract
PURPOSE/AIM Negative feedback controls in endocrine regulatory systems are well recognized. The incretins and their role in glucose regulation have been of major interest recently. Whether the same negative control system applies to the regulation of incretin secretion is not clear. We sought to examine the hypothesis that exogenous administration of glucagon like peptide-1, GLP-1(7-36) amide or its metabolite GLP-1(9-36) amide, reduces the endogenous basal release of this incretin. MATERIALS AND METHODS We evaluated the endogenous basal release of GLP-1 using two separate study designs. In protocol A we examined the GLP-1(7-36) amide levels during the infusion of GLP-1(9-36) amide. In protocol B, we used PYY and GLP-2 as biomarkers for the endogenous basal release of GLP-1(7-36) amide and assessed the endogenous basal release of these two hormones during the GLP-1(7-36) infusion. Twelve lean and 12 obese subjects were enrolled in protocol A and 10 obese volunteers in protocol B. RESULTS The plasma levels of GLP-1(7-36) amide in protocol A and PYY and GLP-2 in protocol B remained unchanged during the exogenous infusion of GLP-1(9-36) and GLP-1(7-36) amide, respectively. CONCLUSIONS The negative feedback control system as described by inhibition of the release of endogenous hormone while infusing it exogenously was not observed for the basal secretion of GLP-1(7-36) amide.
Collapse
Affiliation(s)
| | | | - Olga D. Carlson
- Diabetes Section, National Institute on Aging, Baltimore, MD
| | - Graydon S. Meneilly
- Department of Medicine, University of British Columbia School of Medicine, Vancouver, BC, Canada
| | - Joel F. Habener
- Department of Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, MA
| | | |
Collapse
|
119
|
Palmer BF, Clegg DJ. An Emerging Role of Natriuretic Peptides: Igniting the Fat Furnace to Fuel and Warm the Heart. Mayo Clin Proc 2015; 90:1666-78. [PMID: 26518101 DOI: 10.1016/j.mayocp.2015.08.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/08/2015] [Accepted: 08/13/2015] [Indexed: 01/05/2023]
Abstract
Natriuretic peptides are produced in the heart and have been well characterized for their actions in the cardiovascular system to promote diuresis and natriuresis, thereby contributing to maintenance of extracellular fluid volume and vascular tone. For this review, we scanned the literature using PubMed and MEDLINE using the following search terms: beiging, adipose tissue, natriuretic peptides, obesity, and metabolic syndrome. Articles were selected for inclusion if they represented primary data or review articles published from 1980 to 2015 from high-impact journals. With the advent of the newly approved class of drugs that inhibit the breakdown of natriuretic peptides, thereby increasing their circulation, we highlight additional functions for natriuretic peptides that have recently become appreciated, including their ability to drive lipolysis, facilitate beiging of adipose tissues, and promote lipid oxidation and mitochondrial respiration in skeletal muscle. We provide evidence for new roles for natriuretic peptides, emphasizing their ability to participate in body weight regulation and energy homeostasis and discuss how they may lead to novel strategies to treat obesity and the metabolic syndrome.
Collapse
Affiliation(s)
- Biff F Palmer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Deborah J Clegg
- Biomedical Research Department, Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Beverly Hills, CA.
| |
Collapse
|
120
|
Ramos HR, Birkenfeld AL, de Bold AJ. INTERACTING DISCIPLINES: Cardiac natriuretic peptides and obesity: perspectives from an endocrinologist and a cardiologist. Endocr Connect 2015; 4:R25-36. [PMID: 26115665 PMCID: PMC4485177 DOI: 10.1530/ec-15-0018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [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
Since their discovery in 1981, the cardiac natriuretic peptides (cNP) atrial natriuretic peptide (also referred to as atrial natriuretic factor) and brain natriuretic peptide have been well characterised in terms of their renal and cardiovascular actions. In addition, it has been shown that cNP plasma levels are strong predictors of cardiovascular events and mortality in populations with no apparent heart disease as well as in patients with established cardiac pathology. cNP secretion from the heart is increased by humoral and mechanical stimuli. The clinical significance of cNP plasma levels has been shown to differ in obese and non-obese subjects. Recent lines of evidence suggest important metabolic effects of the cNP system, which has been shown to activate lipolysis, enhance lipid oxidation and mitochondrial respiration. Clinically, these properties lead to browning of white adipose tissue and to increased muscular oxidative capacity. In human association studies in patients without heart disease higher cNP concentrations were observed in lean, insulin-sensitive subjects. Highly elevated cNP levels are generally observed in patients with systolic heart failure or high blood pressure, while obese and type-2 diabetics display reduced cNP levels. Together, these observations suggest that the cNP system plays a role in the pathophysiology of metabolic vascular disease. Understanding this role should help define novel principles in the treatment of cardiometabolic disease.
Collapse
Affiliation(s)
- Hugo R Ramos
- Department of Internal Medicine, Faculty of Medicine, Hospital de Urgencias, National University of Córdoba, Córdoba, X5000,
Argentina
- Correspondence should be addressed to H R Ramos or A L Birkenfeld or
| | - Andreas L Birkenfeld
- Section of Metabolic Vascular Medicine, Medical Clinic III and Paul Langerhans Institute Dresden (PLID), Dresden University School of Medicine, 01307 DresdenGermany
- Division of Diabetes and Nutritional Sciences, King's College London, Rayne Institute, London, SE5 9NU, UK
- Correspondence should be addressed to H R Ramos or A L Birkenfeld or
| | - Adolfo J de Bold
- Cardiovascular Endocrinology Laboratory, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| |
Collapse
|
121
|
Angiotensin II Receptor Blocker Neprilysin Inhibitor (ARNI): New Avenues in Cardiovascular Therapy. High Blood Press Cardiovasc Prev 2015; 22:241-6. [PMID: 26100410 DOI: 10.1007/s40292-015-0112-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/10/2015] [Indexed: 12/11/2022] Open
Abstract
The burden of cardiovascular disease (CVD) is continuously and progressively raising worldwide. Essential hypertension is a major driver of cardiovascular events, including coronary artery disease, myocardial infarction, ischemic stroke and congestive heart failure. This latter may represent the final common pathway of different cardiovascular diseases, and it is often mediated by progressive uncontrolled hypertension. Despite solid advantages derived from effective and sustained blood pressure control, and the widespread availability of effective antihypertensive medications, the vast majority of the more than 1 billion hypertensive patients worldwide continue to have uncontrolled hypertension. Among various factors that may be involved, the abnormal activation of neurohormonal systems is one consistent feature throughout the continuum of cardiovascular diseases. These systems may initiate biologically meaningful "injury responses". However, their sustained chronic overactivity often may induce and maintain the progression from hypertension towards congestive heart failure. The renin-angiotensin-aldosteron system, the sympathetic nervous system and the endothelin system are major neurohormonal stressor systems that are not only able to elevate blood pressure levels by retaining water and sodium, but also to play a role in the pathophysiology of cardiovascular diseases. More recently, the angiotensin receptor neprilysin inhibitor (ARNI) represents a favourable approach to inhibit neutral endopeptidase (NEP) and suppress the RAAS via blockade of the AT1 receptors, without the increased risk of angioedema. LCZ696, the first-in-class ARNI, has already demonstrated BP lowering efficacy in patients with hypertension, in particular with respect to systolic blood pressure levels, improved cardiac biomarkers, cardiac remodelling and prognosis in patients with heart failure. This manuscript will briefly overview the main pathophysiological and therapeutic aspects of ARNI in the clinical management of hypertension and heart failure.
Collapse
|
122
|
Natriuretic peptides in the cross-talk of human cardiovascular and metabolic regulation. J Hypertens 2015; 33:1139-41. [DOI: 10.1097/hjh.0000000000000575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
123
|
Schlueter N, de Sterke A, Willmes DM, Spranger J, Jordan J, Birkenfeld AL. Metabolic actions of natriuretic peptides and therapeutic potential in the metabolic syndrome. Pharmacol Ther 2014; 144:12-27. [PMID: 24780848 DOI: 10.1016/j.pharmthera.2014.04.007] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 04/14/2014] [Indexed: 12/26/2022]
Abstract
Natriuretic peptides (NPs) are a group of peptide-hormones mainly secreted from the heart, signaling via c-GMP coupled receptors. NP are well known for their renal and cardiovascular actions, reducing arterial blood pressure as well as sodium reabsorption. Novel physiological functions have been discovered in recent years, including activation of lipolysis, lipid oxidation, and mitochondrial respiration. Together, these responses promote white adipose tissue browning, increase muscular oxidative capacity, particularly during physical exercise, and protect against diet-induced obesity and insulin resistance. Exaggerated NP release is a common finding in congestive heart failure. In contrast, NP deficiency is observed in obesity and in type-2 diabetes, pointing to an involvement of NP in the pathophysiology of metabolic disease. Based upon these findings, the NP system holds the potential to be amenable to therapeutical intervention against pandemic diseases such as obesity, insulin resistance, and arterial hypertension. Various therapeutic approaches are currently under development. This paper reviews the current knowledge on the metabolic effects of the NP system and discusses potential therapeutic applications.
Collapse
Affiliation(s)
- Nina Schlueter
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité, University School of Medicine, Berlin, Germany
| | - Anita de Sterke
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité, University School of Medicine, Berlin, Germany
| | - Diana M Willmes
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité, University School of Medicine, Berlin, Germany
| | - Joachim Spranger
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité, University School of Medicine, Berlin, Germany
| | - Jens Jordan
- Institute of Clinical Pharmacology, Hannover Medical School, Hannover, Germany
| | - Andreas L Birkenfeld
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité, University School of Medicine, Berlin, Germany.
| |
Collapse
|
124
|
Zraika S, Koh DS, Barrow BM, Lu B, Kahn SE, Andrikopoulos S. Neprilysin deficiency protects against fat-induced insulin secretory dysfunction by maintaining calcium influx. Diabetes 2013; 62:1593-601. [PMID: 23328128 PMCID: PMC3636612 DOI: 10.2337/db11-1593] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Neprilysin contributes to free fatty acid (FFA)-induced cellular dysfunction in nonislet tissues in type 2 diabetes. Here, we show for the first time that with prolonged FFA exposure, islet neprilysin is upregulated and this is associated with reduced insulin pre-mRNA and ATP levels, oxidative/nitrative stress, impaired potassium and calcium channel activities, and decreased glucose-stimulated insulin secretion (GSIS). Genetic ablation of neprilysin specifically protects against FFA-induced impairment of calcium influx and GSIS in vitro and in vivo but does not ameliorate other FFA-induced defects. Importantly, adenoviral overexpression of neprilysin in islets cultured without FFA reproduces the defects in both calcium influx and GSIS, suggesting that upregulation of neprilysin per se mediates insulin secretory dysfunction and that the mechanism for protection conferred by neprilysin deletion involves prevention of reduced calcium influx. Our findings highlight the critical nature of calcium signaling for normal insulin secretion and suggest that interventions to inhibit neprilysin may improve β-cell function in obese humans with type 2 diabetes.
Collapse
Affiliation(s)
- Sakeneh Zraika
- Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, Washington, USA.
| | | | | | | | | | | |
Collapse
|
125
|
Habener JF, Stanojevic V. Alpha cells come of age. Trends Endocrinol Metab 2013; 24:153-63. [PMID: 23260869 DOI: 10.1016/j.tem.2012.10.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 10/27/2012] [Accepted: 10/30/2012] [Indexed: 02/07/2023]
Abstract
The alpha cells that coinhabit the islets with the insulin-producing beta cells have recently captured the attention of diabetes researchers because of new breakthrough findings highlighting the importance of these cells in the maintenance of beta cell health and functions. In normal physiological conditions alpha cells produce glucagon but in conditions of beta cell injury they also produce glucagon-like peptide-1 (GLP-1), a growth and survival factor for beta cells. In this review we consider these new findings on the functions of alpha cells. Alpha cells remain somewhat enigmatic inasmuch as they now appear to be important in the maintenance of the health of beta cells, but their production of glucagon promotes diabetes. This circumstance prompts an examination of approaches to coax alpha cells to produce GLP-1 instead of glucagon.
Collapse
Affiliation(s)
- Joel F Habener
- Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | | |
Collapse
|
126
|
Karoor V, Oka M, Walchak SJ, Hersh LB, Miller YE, Dempsey EC. Neprilysin regulates pulmonary artery smooth muscle cell phenotype through a platelet-derived growth factor receptor-dependent mechanism. Hypertension 2013; 61:921-30. [PMID: 23381789 DOI: 10.1161/hypertensionaha.111.199588] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Reduced neprilysin (NEP), a cell surface metallopeptidase, which cleaves and inactivates proinflammatory and vasoactive peptides, predisposes the lung vasculature to exaggerated remodeling in response to hypoxia. We hypothesize that loss of NEP in pulmonary artery smooth muscle cells results in increased migration and proliferation. Pulmonary artery smooth muscle cells isolated from NEP(-/-) mice exhibited enhanced migration and proliferation in response to serum and platelet-derived growth factor, which was attenuated by NEP replacement. Inhibition of NEP by overexpression of a peptidase dead mutant or knockdown by small interfering RNA in NEP(+/+) cells increased migration and proliferation. Loss of NEP led to an increase in Src kinase activity and phosphorylation of PTEN, resulting in activation of the platelet-derived growth factor receptor (PDGFR). Knockdown of Src kinase with small interfering RNA or inhibition with PP2, a src kinase inhibitor, decreased PDGFR(Y751) phosphorylation and attenuated migration and proliferation in NEP(-/-) smooth muscle cells. NEP substrates, endothelin 1 or fibroblast growth factor 2, increased activation of Src and PDGFR in NEP(+/+) cells, which was decreased by an endothelin A receptor antagonist, neutralizing antibody to fibroblast growth factor 2 and Src inhibitor. Similar to the observations in pulmonary artery smooth muscle cells, levels of phosphorylated PDGFR, Src, and PTEN were elevated in NEP(-/-) lungs. Endothelin A receptor antagonist also attenuated the enhanced responses in NEP(-/-) pulmonary artery smooth muscle cells and lungs. Taken together our results suggest a novel mechanism for the regulation of PDGFR signaling by NEP substrates involving Src and PTEN. Strategies that increase lung NEP activity/expression or target key downstream effectors, like Src, PTEN, or PDGFR, may be of therapeutic benefit in pulmonary vascular disease.
Collapse
Affiliation(s)
- Vijaya Karoor
- Cardiovascular Pulmonary Research Laboratory, RC-2 Room 8118, University of Colorado Anschutz Medical Campus, 12700 E 19th Ave, RC-2, Aurora, CO 80045, USA.
| | | | | | | | | | | |
Collapse
|
127
|
Mangiafico S, Costello-Boerrigter LC, Andersen IA, Cataliotti A, Burnett JC. Neutral endopeptidase inhibition and the natriuretic peptide system: an evolving strategy in cardiovascular therapeutics. Eur Heart J 2012; 34:886-893c. [PMID: 22942338 DOI: 10.1093/eurheartj/ehs262] [Citation(s) in RCA: 196] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hypertension and heart failure (HF) are common diseases that, despite advances in medical therapy, continue to be associated with high morbidity and mortality. Therefore, innovative therapeutic strategies are needed. Inhibition of the neutral endopeptidase (NEPinh) had been investigated as a potential novel therapeutic approach because of its ability to increase the plasma concentrations of the natriuretic peptides (NPs). Indeed, the NPs have potent natriuretic and vasodilator properties, inhibit the activity of the renin-angiotensin-aldosterone system, lower sympathetic drive, and have antiproliferative and antihypertrophic effects. Such potentially beneficial effects can be theoretically achieved by the use of NEPinh. However, studies have shown that NEPinh alone does not result in clinically meaningful blood pressure-lowering actions. More recently, NEPinh has been used in combination with other cardiovascular agents, such as angiotensin-converting enzyme inhibitors, and antagonists of the angiotensin receptor. Another future possible combination would be the use of NEPinh with NPs or their newly developed chimeric peptides. This review summarizes the current knowledge of the use and effects of NEPinh alone or in combination with other therapeutic agents for the treatment of human cardiovascular disease such as HF and hypertension.
Collapse
Affiliation(s)
- Sarah Mangiafico
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Mayo Clinic and Mayo Clinic College of Medicine, Guggenheim 9, 200 First Street SW, Rochester, MN 55901, USA
| | | | | | | | | |
Collapse
|
128
|
Patel VB, Bodiga S, Basu R, Das SK, Wang W, Wang Z, Lo J, Grant MB, Zhong J, Kassiri Z, Oudit GY. Loss of angiotensin-converting enzyme-2 exacerbates diabetic cardiovascular complications and leads to systolic and vascular dysfunction: a critical role of the angiotensin II/AT1 receptor axis. Circ Res 2012; 110:1322-35. [PMID: 22474255 DOI: 10.1161/circresaha.112.268029] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
RATIONALE Diabetic cardiovascular complications are reaching epidemic proportions. Angiotensin-converting enzyme-2 (ACE2) is a negative regulator of the renin-angiotensin system. We hypothesize that loss of ACE2 exacerbates cardiovascular complications induced by diabetes. OBJECTIVE To define the role of ACE2 in diabetic cardiovascular complications. METHODS AND RESULTS We used the well-validated Akita mice, a model of human diabetes, and generated double-mutant mice using the ACE2 knockout (KO) mice (Akita/ACE2(-/y)). Diabetic state was associated with increased ACE2 in Akita mice, whereas additional loss of ACE2 in these mice leads to increased plasma and tissue angiotensin II levels, resulting in systolic dysfunction on a background of impaired diastolic function. Downregulation of SERCA2 and lipotoxicity were equivalent in Akita and Akita/ACE2KO hearts and are likely mediators of the diastolic dysfunction. However, greater activation of protein kinase C and loss of Akt and endothelial nitric oxide synthase phosphorylation occurred in the Akita/ACE2KO hearts. Systolic dysfunction in Akita/ACE2KO mice was linked to enhanced activation of NADPH oxidase and metalloproteinases, resulting in greater oxidative stress and degradation of the extracellular matrix. Impaired flow-mediated dilation in vivo correlated with increased vascular oxidative stress in Akita/ACE2KO mice. Treatment with the AT1 receptor blocker, irbesartan rescued the systolic dysfunction, normalized altered signaling pathways, flow-mediated dilation, and the increased oxidative stress in the cardiovascular system. CONCLUSIONS Loss of ACE2 disrupts the balance of the renin-angiotensin system in a diabetic state and leads to an angiotensin II/AT1 receptor-dependent systolic dysfunction and impaired vascular function. Our study demonstrates that ACE2 serves as a protective mechanism against diabetes-induced cardiovascular complications.
Collapse
Affiliation(s)
- Vaibhav B Patel
- Division of Cardiology, Department of Medicine, Mazankowski Alberta Heart Institute, University of Alberta, Alberta, Edmonton, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Nakatsuji H, Kishida K, Funahashi T, Nakagawa T, Shimomura I. Hyperinsulinemia correlates with low levels of plasma B-type natriuretic peptide in Japanese men irrespective of fat distribution. Cardiovasc Diabetol 2012; 11:22. [PMID: 22397400 PMCID: PMC3320543 DOI: 10.1186/1475-2840-11-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 03/07/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND B-type natriuretic peptide (BNP), a member of the natriuretic peptide family, is a cardiac-derived secretory hormone with natriuretic, diuretic, and vasorelaxant activities. Intraabdominal fat accumulation is associated with atherosclerotic cardiovascular diseases and cardiac dysfunction. Circulating BNP levels are relatively low (within the normal limits) in obesity and the metabolic syndrome. However, the relationship between plasma BNP levels and visceral fat accumulation in general population has not been reported. The present study analyzed the relationships between plasma BNP levels and various clinical variables, including insulin, visceral and subcutaneous fat area (VFA and SFA, respectively), in normal Japanese men. METHODS The study (Victor-J study) subjects were consecutive 500 Japanese male workers, who underwent a health checkup and were measured VFA and SFA by computed tomography. RESULTS Age-adjusted simple linear regression analysis showed that log-BNP correlated positively with HDL-cholesterol, and negatively with VFA, log-immunoreactive insulin (IRI), log-triglyceride, and LDL-cholesterol, but not body mass index or SFA. Stepwise multiple regression analysis identified log-IRI and HDL-cholesterol as significant determinants of log-BNP. Subjects with IRI ≥5.5 μIU/mL had lower plasma BNP levels than those with IRI < 5.5 μIU/mL, irrespective of obesity (body mass index, cutoff value 25 kg/m2), visceral fat accumulation (VFA, cutoff value 100 cm2) and subcutaneous fat accumulation (SFA, cutoff value 128 cm2). CONCLUSIONS Our study showed that hyperinsulinemia correlated with low levels of plasma BNP in general men, irrespective of fat distribution. TRIAL REGISTRATION UMIN 000004318.
Collapse
Affiliation(s)
- Hideaki Nakatsuji
- Department of Metabolic Medicine, Osaka University, Suita, Osaka, Japan
| | | | | | | | | |
Collapse
|
130
|
Coppey L, Lu B, Gerard C, Yorek MA. Effect of Inhibition of Angiotensin-Converting Enzyme and/or Neutral Endopeptidase on Neuropathy in High-Fat-Fed C57Bl/6J Mice. J Obes 2012; 2012:326806. [PMID: 23056927 PMCID: PMC3465928 DOI: 10.1155/2012/326806] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 08/14/2012] [Accepted: 08/28/2012] [Indexed: 11/17/2022] Open
Abstract
We have demonstrated that treating diet-induced obese (DIO) mice with the vasopeptidase inhibitor ilepatril improved neural function. Vasopeptidase inhibitors block angiotensin-converting enzyme (ACE) and neutral endopeptidase (NEP) activity. We propose that increased activity of ACE and NEP contributes to pathophysiology of DIO. To address this issue C57Bl/6J mice or mice deficient in NEP were fed a high-fat diet and treated with ilepatril, enalapril, ACE inhibitor, or candoxatril, NEP inhibitor, using both prevention and intervention protocols. Endpoints included glucose utilization and neural function determination. In the prevention study glucose tolerance was impaired in DIO C57Bl/6J mice and improved with ilepatril or enalapril. Sensory nerve conduction velocity, thermal nociception, and intraepidermal nerve fiber density were impaired in DIO C57Bl/6J mice and improved with ilepatril or candoxatril. In the intervention study only enalapril improved glucose tolerance. Sensory nerve conduction velocity and intraepidermal nerve fiber density were improved by all three treatments, whereas thermal nociception was improved by ilepatril or candoxatril. In NEP-deficient mice DIO impaired glucose utilization and this was improved with enalapril. Nerve function was not impaired by DIO in NEP-deficient mice. These studies suggest that ACE and NEP play a role in pathophysiology associated with DIO.
Collapse
Affiliation(s)
- Lawrence Coppey
- Department of Veterans Affairs Iowa City Health Care System, Iowa City, IA 52246, USA
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Bao Lu
- Ina Sue Perlmutter Laboratory, Children's Hospital, Department of Pediatrics and Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Craig Gerard
- Ina Sue Perlmutter Laboratory, Children's Hospital, Department of Pediatrics and Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Mark A. Yorek
- Department of Veterans Affairs Iowa City Health Care System, Iowa City, IA 52246, USA
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52246, USA
- *Mark A. Yorek:
| |
Collapse
|
131
|
Human matrix metalloproteinases: an ubiquitarian class of enzymes involved in several pathological processes. Mol Aspects Med 2011; 33:119-208. [PMID: 22100792 DOI: 10.1016/j.mam.2011.10.015] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 10/29/2011] [Indexed: 02/07/2023]
Abstract
Human matrix metalloproteinases (MMPs) belong to the M10 family of the MA clan of endopeptidases. They are ubiquitarian enzymes, structurally characterized by an active site where a Zn(2+) atom, coordinated by three histidines, plays the catalytic role, assisted by a glutamic acid as a general base. Various MMPs display different domain composition, which is very important for macromolecular substrates recognition. Substrate specificity is very different among MMPs, being often associated to their cellular compartmentalization and/or cellular type where they are expressed. An extensive review of the different MMPs structural and functional features is integrated with their pathological role in several types of diseases, spanning from cancer to cardiovascular diseases and to neurodegeneration. It emerges a very complex and crucial role played by these enzymes in many physiological and pathological processes.
Collapse
|
132
|
Kassi E, Pervanidou P, Kaltsas G, Chrousos G. Metabolic syndrome: definitions and controversies. BMC Med 2011; 9:48. [PMID: 21542944 PMCID: PMC3115896 DOI: 10.1186/1741-7015-9-48] [Citation(s) in RCA: 900] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 05/05/2011] [Indexed: 01/19/2023] Open
Abstract
Metabolic syndrome (MetS) is a complex disorder defined by a cluster of interconnected factors that increase the risk of cardiovascular atherosclerotic diseases and diabetes mellitus type 2. Currently, several different definitions of MetS exist, causing substantial confusion as to whether they identify the same individuals or represent a surrogate of risk factors. Recently, a number of other factors besides those traditionally used to define MetS that are also linked to the syndrome have been identified. In this review, we critically consider existing definitions and evolving information, and conclude that there is still a need to develop uniform criteria to define MetS, so as to enable comparisons between different studies and to better identify patients at risk. As the application of the MetS model has not been fully validated in children and adolescents as yet, and because of its alarmingly increasing prevalence in this population, we suggest that diagnosis, prevention and treatment in this age group should better focus on established risk factors rather than the diagnosis of MetS.
Collapse
Affiliation(s)
- Eva Kassi
- Department of Biochemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiota Pervanidou
- First Department of Paediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Gregory Kaltsas
- Department of Pathophysiology, National and Kapodistrian University of Athens, Athens, Greece
| | - George Chrousos
- First Department of Paediatrics, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|