Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Dev Cell ; 52(6): 748-763.e6, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32109384

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a multisystemic genetic disorder caused by the CTG repeat expansion in the 3'-untranslated region of DMPK gene. Heart dysfunctions occur in ∼80% of DM1 patients and are the second leading cause of DM1-related deaths. Herein, we report that upregulation of a non-muscle splice isoform of RNA-binding protein RBFOX2 in DM1 heart tissue-due to altered splicing factor and microRNA activities-induces cardiac conduction defects in DM1 individuals. Mice engineered to express the non-muscle RBFOX240 isoform in heart via tetracycline-inducible transgenesis, or CRISPR/Cas9-mediated genome editing, reproduced DM1-related cardiac conduction delay and spontaneous episodes of arrhythmia. Further, by integrating RNA binding with cardiac transcriptome datasets from DM1 patients and mice expressing the non-muscle RBFOX2 isoform, we identified RBFOX240-driven splicing defects in voltage-gated sodium and potassium channels, which alter their electrophysiological properties. Thus, our results uncover a trans-dominant role for an aberrantly expressed RBFOX240 isoform in DM1 cardiac pathogenesis.


Subject(s)
Action Potentials , Heart Rate , Myotonic Dystrophy/genetics , RNA Splicing Factors/genetics , RNA Splicing , Repressor Proteins/genetics , Adult , Animals , Cells, Cultured , Female , Humans , Male , Mice, Inbred C57BL , Mice, Inbred ICR , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/physiopathology , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing Factors/metabolism , Repressor Proteins/metabolism , Voltage-Gated Sodium Channels/genetics , Voltage-Gated Sodium Channels/metabolism
4.
J Clin Invest ; 129(8): 3171-3184, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31264976

ABSTRACT

Arrhythmogenic cardiomyopathy (ACM) is an inherited arrhythmia syndrome characterized by severe structural and electrical cardiac phenotypes, including myocardial fibrofatty replacement and sudden cardiac death. Clinical management of ACM is largely palliative, owing to an absence of therapies that target its underlying pathophysiology, which stems partially from our limited insight into the condition. Following identification of deceased ACM probands possessing ANK2 rare variants and evidence of ankyrin-B loss of function on cardiac tissue analysis, an ANK2 mouse model was found to develop dramatic structural abnormalities reflective of human ACM, including biventricular dilation, reduced ejection fraction, cardiac fibrosis, and premature death. Desmosomal structure and function appeared preserved in diseased human and murine specimens in the presence of markedly abnormal ß-catenin expression and patterning, leading to identification of a previously unknown interaction between ankyrin-B and ß-catenin. A pharmacological activator of the WNT/ß-catenin pathway, SB-216763, successfully prevented and partially reversed the murine ACM phenotypes. Our findings introduce what we believe to be a new pathway for ACM, a role of ankyrin-B in cardiac structure and signaling, a molecular link between ankyrin-B and ß-catenin, and evidence for targeted activation of the WNT/ß-catenin pathway as a potential treatment for this disease.


Subject(s)
Ankyrins , Arrhythmogenic Right Ventricular Dysplasia , Myocardium , Wnt Signaling Pathway , Animals , Ankyrins/genetics , Ankyrins/metabolism , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/metabolism , Arrhythmogenic Right Ventricular Dysplasia/pathology , Disease Models, Animal , Female , Humans , Indoles/pharmacology , Male , Maleimides/pharmacology , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , beta Catenin/genetics , beta Catenin/metabolism
5.
J Biol Chem ; 294(24): 9576-9591, 2019 06 14.
Article in English | MEDLINE | ID: mdl-31064843

ABSTRACT

Spectrins are cytoskeletal proteins essential for membrane biogenesis and regulation and serve critical roles in protein targeting and cellular signaling. αII spectrin (SPTAN1) is one of two α spectrin genes and αII spectrin dysfunction is linked to alterations in axon initial segment formation, cortical lamination, and neuronal excitability. Furthermore, human αII spectrin loss-of-function variants cause neurological disease. As global αII spectrin knockout mice are embryonic lethal, the in vivo roles of αII spectrin in adult heart are unknown and untested. Here, based on pronounced alterations in αII spectrin regulation in human heart failure we tested the in vivo roles of αII spectrin in the vertebrate heart. We created a mouse model of cardiomyocyte-selective αII spectrin-deficiency (cKO) and used this model to define the roles of αII spectrin in cardiac function. αII spectrin cKO mice displayed significant structural, cellular, and electrical phenotypes that resulted in accelerated structural remodeling, fibrosis, arrhythmia, and mortality in response to stress. At the molecular level, we demonstrate that αII spectrin plays a nodal role for global cardiac spectrin regulation, as αII spectrin cKO hearts exhibited remodeling of αI spectrin and altered ß-spectrin expression and localization. At the cellular level, αII spectrin deficiency resulted in altered expression, targeting, and regulation of cardiac ion channels NaV1.5 and KV4.3. In summary, our findings define critical and unexpected roles for the multifunctional αII spectrin protein in the heart. Furthermore, our work provides a new in vivo animal model to study the roles of αII spectrin in the cardiomyocyte.


Subject(s)
Arrhythmias, Cardiac/pathology , Disease Models, Animal , Heart Failure/pathology , Ischemia/pathology , Myocytes, Cardiac/pathology , Spectrin/physiology , Animals , Arrhythmias, Cardiac/etiology , Cells, Cultured , Female , Heart Failure/etiology , Humans , Ischemia/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
6.
Circ Res ; 124(5): 737-746, 2019 03.
Article in English | MEDLINE | ID: mdl-30602331

ABSTRACT

RATIONALE: Voltage-gated Na+ channel ( INa) function is critical for normal cardiac excitability. However, the Na+ channel late component ( INa,L) is directly associated with potentially fatal forms of congenital and acquired human arrhythmia. CaMKII (Ca2+/calmodulin-dependent kinase II) enhances INa,L in response to increased adrenergic tone. However, the pathways that negatively regulate the CaMKII/Nav1.5 axis are unknown and essential for the design of new therapies to regulate the pathogenic INa,L. OBJECTIVE: To define phosphatase pathways that regulate INa,L in vivo. METHODS AND RESULTS: A mouse model lacking a key regulatory subunit (B56α) of the PP (protein phosphatase) 2A holoenzyme displayed aberrant action potentials after adrenergic stimulation. Unbiased computational modeling of B56α KO (knockout) mouse myocyte action potentials revealed an unexpected role of PP2A in INa,L regulation that was confirmed by direct INa,L recordings from B56α KO myocytes. Further, B56α KO myocytes display decreased sensitivity to isoproterenol-induced induction of arrhythmogenic INa,L, and reduced CaMKII-dependent phosphorylation of Nav1.5. At the molecular level, PP2A/B56α complex was found to localize and coimmunoprecipitate with the primary cardiac Nav channel, Nav1.5. CONCLUSIONS: PP2A regulates Nav1.5 activity in mouse cardiomyocytes. This regulation is critical for pathogenic Nav1.5 late current and requires PP2A-B56α. Our study supports B56α as a novel target for the treatment of arrhythmia.


Subject(s)
Arrhythmias, Cardiac/enzymology , Heart Rate , Ion Channel Gating , Myocytes, Cardiac/enzymology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Protein Phosphatase 2/metabolism , Action Potentials , Adrenergic beta-Agonists/pharmacology , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Female , Genetic Predisposition to Disease , Humans , Ion Channel Gating/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Phenotype , Phosphorylation , Protein Phosphatase 2/deficiency , Protein Phosphatase 2/genetics , Time Factors
7.
Front Physiol ; 9: 1482, 2018.
Article in English | MEDLINE | ID: mdl-30405438

ABSTRACT

Global obesity rates have nearly tripled since 1975. This obesity rate increase is mirrored by increases in atrial fibrillation (AF) that now impacts nearly 10% of Americans over the age of 65. Numerous epidemiologic studies have linked incidence of AF and obesity and other obesity-related diseases, including hypertension and diabetes. Due to the wealth of epidemiologic data linking AF with obesity-related disease, mechanisms of AF pathogenesis in the context of obesity are an area of ongoing investigation. However, progress has been somewhat slowed by the complex phenotype of obesity; separating the effects of obesity from those of related sequelae is problematic. While the initiation of pathogenic pathways leading to AF varies with disease (including increased glycosylation in diabetes, increased renin angiotensin aldosterone system activation in hypertension, atrial ischemia in coronary artery disease, and sleep apnea) the pathogenesis of AF is united by shared mediators of altered conduction in the atria. We suggest focusing on these downstream mediators of AF in obesity is likely to yield more broadly applicable data. In the context of obesity, AF is driven by the interrelated processes of inflammation, atrial remodeling, and oxidative stress. Obesity is characterized by a constant low-grade inflammation that leads to increased expression of pro-inflammatory cytokines. These cytokines contribute to changes in cardiomyocyte excitability. Atrial structural remodeling, including fibrosis, enlargement, and fatty infiltration is a prominent feature of AF and contributes to the altered conduction. Finally, obesity impacts oxidative stress. Within the cardiomyocyte, oxidative stress is increased through both increased production of reactive oxygen species and by downregulation of scavenging enzymes. This increased oxidative stress modulates of cardiomyocyte excitability, increasing susceptibility to AF. Although the initiating insults vary, inflammation, atrial remodeling, and oxidative stress are conserved mechanisms in the pathophysiology of AF in the obese patients. In this review, we highlight mechanisms that have been shown to be relevant in the pathogenesis of AF across obesity-related disease.

8.
JACC Basic Transl Sci ; 3(5): 675-689, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30456339

ABSTRACT

Ankyrin polypeptides are intracellular proteins responsible for targeting cardiac membrane proteins. Here, the authors demonstrate that ankyrin-G plays an unexpected role in normal compensatory physiological remodeling in response to myocardial stress and aging; the authors implicate disruption of ankyrin-G in human heart failure. Mechanistically, the authors illustrate that ankyrin-G serves as a key nodal protein required for cardiac myofilament integration with the intercalated disc. Their data define novel in vivo mechanistic roles for ankyrin-G, implicate ankyrin-G as necessary for compensatory cardiac physiological remodeling under stress, and implicate disruption of ankyrin-G in the development and progression of human heart failure.

10.
J Mol Cell Cardiol ; 101: 127-133, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27832939

ABSTRACT

Reversible protein phosphorylation is central to a variety of cardiac processes including excitation-contraction coupling, Ca2+ handling, cell metabolism, myofilament regulation, and cell-cell communication. While kinase pathways linked with elevated adrenergic signaling have been a major focus for the cardiovascular field over the past half century, new findings support the critical role of protein phosphatases in both health and disease. Protein phosphatase 2A (PP2A) is a central cardiac phosphatase that regulates diverse myocyte functions through a host of target molecules. Notably, multiple mechanisms have evolved to dynamically tune PP2A function, including modulation of the composition, phosphorylation, methylation, and localization of PP2A holoenzyme populations. Further, aberrations in this regulation of PP2A function may contribute to cardiac pathophysiology. In summary, PP2A is a critical regulatory molecule in both health and disease, with a myriad of targets in heart. Based on their unique structure, localization, and regulatory properties, PP2A subunits represent exciting therapeutic targets to modulate altered adrenergic signaling in cardiovascular disease.


Subject(s)
Myocardium/metabolism , Protein Phosphatase 2/metabolism , Animals , Gene Expression Regulation , Heart Diseases/drug therapy , Heart Diseases/etiology , Heart Diseases/metabolism , Holoenzymes , Humans , Isoenzymes , Molecular Targeted Therapy , Multigene Family , Myocardial Contraction , Protein Binding , Protein Interaction Domains and Motifs , Protein Kinases/metabolism , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/genetics , Protein Processing, Post-Translational , RNA Interference , Signal Transduction
11.
Heart Rhythm ; 13(9): 1932-40, 2016 09.
Article in English | MEDLINE | ID: mdl-27298202

ABSTRACT

BACKGROUND: Human ANK2 (ankyrin-B) loss-of-function variants are directly linked with arrhythmia phenotypes. However, in atypical non-ion channel arrhythmia genes such as ANK2 that lack the same degree of robust structure/function and clinical data, it may be more difficult to assign variant disease risk based simply on variant location, minor allele frequency, and/or predictive structural algorithms. The human ankyrin-B p.L1622I variant found in arrhythmia probands displays significant diversity in minor allele frequency across populations. OBJECTIVE: The objective of this study was to directly test the in vivo impact of ankyrin-B p.L1622I on cardiac electrical phenotypes and arrhythmia risk using a new animal model. METHODS: We tested arrhythmia phenotypes in a new "knock-in" animal model harboring the human ankyrin-B p.L1622I variant. RESULTS: Ankyrin-B p.L1622I displays reduced posttranslational expression in vivo, resulting in reduced cardiac ankyrin-B expression and reduced association with binding-partner Na/Ca exchanger. Ankyrin-B(L1622I/L1622I) mice display changes in heart rate, atrioventricular and intraventricular conduction, and alterations in repolarization. Furthermore, ankyrin-B(L1622I/L1622I) mice display catecholamine-dependent arrhythmias. At the cellular level, ankyrin-B(L1622I/L1622I) myocytes display increased action potential duration and severe arrhythmogenic afterdepolarizations that provide a mechanistic rationale for the arrhythmias. CONCLUSION: Our findings support in vivo arrhythmogenic phenotypes of an ANK2 variant with unusual frequency in select populations. On the basis of our findings and current clinical data, we support classification of p.L1622I as a "mild" loss-of-function variant that may confer arrhythmia susceptibility in the context of secondary risk factors including environment, medication, and/or additional genetic variation.


Subject(s)
Ankyrins/genetics , Arrhythmias, Cardiac/genetics , Action Potentials/genetics , Animals , Arrhythmias, Cardiac/ethnology , Arrhythmias, Cardiac/physiopathology , Black People/genetics , Disease Models, Animal , Gene-Environment Interaction , Genetic Predisposition to Disease/ethnology , Genetic Variation , Humans , Loss of Function Mutation , Mice , Mice, Inbred C57BL , Phenotype , Risk Assessment/ethnology , Risk Factors
12.
Gerontology ; 62(2): 163-72, 2016.
Article in English | MEDLINE | ID: mdl-26372907

ABSTRACT

BACKGROUND: Growth hormone (GH)-resistant/deficient mice experience improved glucose homeostasis and substantially increased lifespan. Recent evidence suggests that long-lived GH-resistant/deficient mice are protected from white adipose tissue (WAT) dysfunction, including WAT cellular senescence, impaired adipogenesis and loss of subcutaneous WAT in old age. This preservation of WAT function has been suggested to be a potential mechanism for the extended lifespan of these mice. OBJECTIVE: The objective of this study was to examine WAT senescence, WAT distribution and glucose homeostasis in dwarf GH receptor antagonist (GHA) transgenic mice, a unique mouse strain having decreased GH action but normal longevity. METHODS: 18-month-old female GHA mice and wild-type (WT) littermate controls were used. Prior to dissection, body composition, fasting blood glucose as well as glucose and insulin tolerance tests were performed. WAT distribution was determined by weighing four distinct WAT depots at the time of dissection. Cellular senescence in four WAT depots was assessed using senescence-associated ß-galactosidase staining to quantify the senescent cell burden, and real-time qPCR to quantify gene expression of senescence markers p16 and IL-6. RESULTS: GHA mice had a 22% reduction in total body weight, a 33% reduction in lean mass and a 10% increase in body fat percentage compared to WT controls. GHA mice had normal fasting blood glucose and improved insulin sensitivity; however, they exhibited impaired glucose tolerance. Moreover, GHA mice displayed enhanced lipid storage in the inguinal subcutaneous WAT depot (p < 0.05) and a 1.7-fold increase in extra-/intraperitoneal WAT ratio compared to controls (p < 0.05). Measurements of WAT cellular senescence showed no difference between GHA mice and WT controls. CONCLUSIONS: Similar to other mice with decreased GH action, female GHA mice display reduced age-related lipid redistribution and improved insulin sensitivity, but no change in cellular senescence. The similar abundance of WAT senescent cells in GHA and control mice suggests that any protection against generation of senescent cells afforded by decreased GH action, low insulin-like growth factor 1 and/or improved insulin sensitivity in the GHA mice may be offset by their severe adiposity, since obesity is known to increase senescence.


Subject(s)
Adipocytes, White , Blood Glucose/metabolism , Cellular Senescence/genetics , Receptors, Somatotropin/genetics , Subcutaneous Fat , Adipogenesis/genetics , Animals , Female , Glucose Intolerance/metabolism , Homeostasis/genetics , Insulin Resistance , Insulin-Like Growth Factor I/metabolism , Laron Syndrome , Mice , Mice, Transgenic , beta-Galactosidase/metabolism
13.
Proc Natl Acad Sci U S A ; 112(46): E6301-10, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26578790

ABSTRACT

Chronic, low grade, sterile inflammation frequently accompanies aging and age-related diseases. Cellular senescence is associated with the production of proinflammatory chemokines, cytokines, and extracellular matrix (ECM) remodeling proteases, which comprise the senescence-associated secretory phenotype (SASP). We found a higher burden of senescent cells in adipose tissue with aging. Senescent human primary preadipocytes as well as human umbilical vein endothelial cells (HUVECs) developed a SASP that could be suppressed by targeting the JAK pathway using RNAi or JAK inhibitors. Conditioned medium (CM) from senescent human preadipocytes induced macrophage migration in vitro and inflammation in healthy adipose tissue and preadipocytes. When the senescent cells from which CM was derived had been treated with JAK inhibitors, the resulting CM was much less proinflammatory. The administration of JAK inhibitor to aged mice for 10 wk alleviated both adipose tissue and systemic inflammation and enhanced physical function. Our findings are consistent with a possible contribution of senescent cells and the SASP to age-related inflammation and frailty. We speculate that SASP inhibition by JAK inhibitors may contribute to alleviating frailty. Targeting the JAK pathway holds promise for treating age-related dysfunction.


Subject(s)
Adipocytes/enzymology , Cellular Senescence/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Janus Kinases/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Adipocytes/cytology , Adipose Tissue/cytology , Adipose Tissue/enzymology , Animals , Cell Movement/drug effects , Cell Movement/genetics , Cellular Senescence/genetics , Extracellular Matrix/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Humans , Janus Kinases/genetics , Janus Kinases/metabolism , Macrophages/cytology , Macrophages/enzymology , Mice , RNA, Small Interfering/genetics , Signal Transduction/genetics
14.
J Endocrinol ; 226(1): 13-23, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25943560

ABSTRACT

GH influences adipocyte differentiation, but both stimulatory and inhibitory effects have been described. Adipose tissue-derived mesenchymal stem cells (AT-MSCs) are multipotent and are able to differentiate into adipocytes, among other cells. Canonical Wnt/ß-catenin signaling activation impairs adipogenesis. The aim of the present study was to elucidate the role of GH on AT-MSC adipogenesis using cells isolated from male GH receptor knockout (GHRKO), bovine GH transgenic (bGH) mice, and wild-type littermate control (WT) mice. AT-MSCs from subcutaneous (sc), epididiymal (epi), and mesenteric (mes) AT depots were identified and isolated by flow cytometry (Pdgfrα+ Sca1+ Cd45- Ter119- cells). Their in vitro adipogenic differentiation capacity was determined by cell morphology and real-time RT-PCR. Using identical in vitro conditions, adipogenic differentiation of AT-MSCs was only achieved in the sc depot, and not in epi and mes depots. Notably, we observed an increased differentiation in cells isolated from sc-GHRKO and an impaired differentiation of sc-bGH cells as compared to sc-WT cells. Axin2, a marker of Wnt/ß-catenin activation, was increased in mature sc-bGH adipocytes, which suggests that activation of this pathway may be responsible for the decreased adipogenesis. Thus, the present study demonstrates that (i) adipose tissue in mice has a well-defined population of Pdgfrα+ Sca1+ MSCs; (ii) the differentiation capacity of AT-MSCs varies from depot to depot regardless of GH genotype; (iii) the lack of GH action increases adipogenesis in the sc depot; and iv) activation of the Wnt/ß-catenin pathway might mediate the GH effect on AT-MSCs. Taken together, the present results suggest that GH diminishes fat mass in part by altering adipogenesis of MSCs.


Subject(s)
Adipogenesis/physiology , Adipose Tissue, White/cytology , Growth Hormone/physiology , Mesenchymal Stem Cells/cytology , Adipocytes/cytology , Animals , Antigens, Ly/metabolism , Cattle , Cell Differentiation , Growth Hormone/genetics , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptors, Somatotropin/deficiency , Receptors, Somatotropin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
16.
Endocrinology ; 156(2): 555-64, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25406017

ABSTRACT

Reduced GH levels have been associated with improved glucose metabolism and increased longevity despite obesity in multiple mouse lines. However, one mouse line, the GH receptor antagonist (GHA) transgenic mouse, defies this trend because it has reduced GH action and increased adiposity, but glucose metabolism and life span are similar to controls. Slight differences in glucose metabolism and adiposity profiles can become exaggerated on a high-fat (HF) diet. Thus, in this study, male and female GHA and wild-type (WT) mice in a C57BL/6 background were placed on HF and low-fat (LF) diets for 11 weeks, starting at 10 weeks of age, to assess how GHA mice respond to additional metabolic stress of HF feeding. On a HF diet, all mice showed significant weight gain, although GHA gained weight more dramatically than WT mice, with males gaining more than females. Most of this weight gain was due to an increase in fat mass with WT mice increasing primarily in the white adipose tissue perigonadal depots, whereas GHA mice gained in both the sc and perigonadal white adipose tissue regions. Notably, GHA mice were somewhat protected from detrimental glucose metabolism changes on a HF diet because they had only modest increases in serum glucose levels, remained glucose tolerant, and did not develop hyperinsulinemia. Sex differences were observed in many measures with males reacting more dramatically to both a reduction in GH action and HF diet. In conclusion, our findings show that GHA mice, which are already obese, are susceptible to further adipose tissue expansion with HF feeding while remaining resilient to alterations in glucose homeostasis.


Subject(s)
Diet, High-Fat/adverse effects , Obesity/etiology , Receptors, Somatotropin/antagonists & inhibitors , Adipocytes/pathology , Adiposity , Animals , Body Weight , Cattle , Disease Models, Animal , Eating , Female , Glucose/metabolism , Glucose Intolerance , Hyperinsulinism , Insulin/blood , Leptin/blood , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Obesity/blood , Obesity/pathology , Receptors, Somatotropin/metabolism , Triglycerides/metabolism , Weight Gain
17.
Endocrinology ; 156(5): 1794-803, 2015 May.
Article in English | MEDLINE | ID: mdl-25521584

ABSTRACT

White adipose tissue (WAT) is composed of mature adipocytes and a stromal vascular fraction (SVF), which contains a variety of cells, including immune cells that vary among the different WAT depots. Growth hormone (GH) impacts immune function and adiposity in an adipose depot-specific manner. However, its effects on WAT immune cell populations remain unstudied. Bovine GH transgenic (bGH) mice are commonly used to study the in vivo effects of GH. These giant mice have an excess of GH action, impaired glucose metabolism, decreased adiposity, increased lean mass, and a shortened lifespan. Therefore, the purpose of this study was to characterize the WAT depot-specific differences in immune cell populations in the presence of excess GH in vivo. Three WAT depots were assessed: inguinal (sc), epididymal (EPI), and mesenteric (MES). Subcutaneous and MES bGH WAT depots showed a significantly higher number of total SVF cells, yet only MES bGH WAT had higher leukocyte counts compared with control samples. By means of flow cytometry analysis of the SVF, we detected greater macrophage and regulatory T-cell infiltration in sc and MES bGH WAT depots compared with controls. However, no differences were observed in the EPI WAT depot. RNA-sequencing confirmed significant alterations in pathways related to T-cell infiltration and activation in the sc depot with fewer significant changes in the EPI bGH WAT depot. These findings collectively point to a previously unrecognized role for GH in influencing the distribution of WAT immune cell populations in a depot-specific manner.


Subject(s)
Adipose Tissue, White/immunology , Growth Hormone/genetics , Macrophages/immunology , Stromal Cells/cytology , T-Lymphocytes, Regulatory/immunology , Adipose Tissue, White/cytology , Adiposity/immunology , Animals , Cattle , Growth Hormone/immunology , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/immunology , Leukocyte Count , Macrophages/cytology , Male , Mice , Mice, Transgenic , Subcutaneous Fat/cytology , Subcutaneous Fat/immunology
18.
Aging (Albany NY) ; 6(7): 575-86, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25063774

ABSTRACT

The aging process is associated with the development of several chronic diseases. White adipose tissue (WAT) may play a central role in age-related disease onset and progression due to declines in adipogenesis with advancing age. Recent reports indicate that the accumulation of senescent progenitor cells may be involved in age-related WAT dysfunction. Growth hormone (GH) action has profound effects on adiposity and metabolism and is known to influence lifespan. In the present study we tested the hypothesis that GH activity would predict age-related WAT dysfunction and accumulation of senescent cells. We found that long-lived GH-deficient and -resistant mice have reduced age-related lipid redistribution. Primary preadipocytes from GH-resistant mice also were found to have greater differentiation capacity at 20 months of age when compared to controls. GH activity was also found to be positively associated with senescent cell accumulation in WAT. Our results demonstrate an association between GH activity, age-related WAT dysfunction, and WAT senescent cell accumulation in mice. Further studies are needed to determine if GH is directly inducing cellular senescence in WAT or if GH actions on other target organs or alternative downstream alterations in insulin-like growth factor-1, insulin or glucose levels are responsible.


Subject(s)
Adipocytes/physiology , Adipose Tissue, White/physiopathology , Aging/physiology , Cellular Senescence/physiology , Growth Hormone/pharmacology , Adipose Tissue, White/cytology , Animals , Cell Differentiation , Lipid Metabolism , Mice , Mice, Inbred Strains , Mutation , RNA, Messenger
19.
J Gerontol A Biol Sci Med Sci ; 69(2): 131-41, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23695394

ABSTRACT

Reduced growth hormone (GH) action is associated with extended longevity in many vertebrate species. GH receptor (GHR) null (GHR(-)(/-)) mice, which have a disruption in the GHR gene, are a well-studied example of mice that are insulin sensitive and long lived yet obese. However, unlike other mouse lines with reduced GH action, GH receptor antagonist (GHA) transgenic mice have reduced GH action yet exhibit a normal, not extended, life span. Understanding why GHA mice do not have extended life span though they share many physiological attributes with GHR(-)(/-) mice will help provide clues about how GH influences aging. For this study, we examined age- and sex-related changes in body composition, glucose homeostasis, circulating adipokines, and tissue weights in GHA mice and littermate controls. Compared with previous studies with GHR(-)(/-) mice, GHA mice had more significant increases in fat mass with advancing age. The increased obesity resulted in significant adipokine changes. Euglycemia was maintained in GHA mice; however, hyperinsulinemia developed in older male GHA mice. Overall, GHA mice experience a more substantial, generalized obesity accompanied by altered adipokine levels and glucose homeostasis than GHR(-)(/-) mice, which becomes more exaggerated with advancing age and which likely contributes to the lack of life-span extension in these mice.


Subject(s)
Adiposity/physiology , Growth Hormone/physiology , Insulin-Like Growth Factor I/physiology , Insulin/physiology , Leptin/physiology , Longevity/physiology , Adiponectin/metabolism , Animals , Blood Glucose/metabolism , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic , Receptors, Somatotropin/antagonists & inhibitors , Sex Factors
20.
J Gerontol A Biol Sci Med Sci ; 69(1): 34-43, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23873966

ABSTRACT

Growth hormone receptor-null (GHR(-/-)) mice are dwarf, insulin sensitive, and long-lived in spite of increased adiposity. However, their adiposity is not uniform, with select white adipose tissue (WAT) depots enlarged. To study WAT depot-specific effects on insulin sensitivity and life span, we analyzed individual WAT depots of 12- and 24-month-old GHR(-) (/-) and wild-type (WT) mice, as well as their plasma levels of selected hormones. Adipocyte sizes and plasma insulin, leptin, and adiponectin levels decreased with age in both GHR(-) (/-) and WT mice. Two-dimensional gel electrophoresis proteomes of WAT depots were similar among groups, but several proteins involved in endocytosis and/or cytoskeletal organization (Ehd2, S100A10, actin), anticoagulation (S100A10, annexin A5), and age-related conditions (alpha2-macroglobulin, apolipoprotein A-I, transthyretin) showed significant differences between genotypes. Because Ehd2 may regulate endocytosis of Glut4, we measured Glut4 levels in the WAT depots of GHR(-) (/-) and WT mice. Inguinal WAT of 12-month-old GHR(-) (/-) mice displayed lower levels of Glut4 than WT. Overall, the protein changes detected in this study offer new insights into possible mechanisms contributing to enhanced insulin sensitivity and extended life span in GHR(-) (/-) mice.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, White/metabolism , Aging/physiology , DNA/genetics , Gene Expression Regulation, Developmental , Insulin Resistance , Receptors, Somatotropin/genetics , Adipocytes/cytology , Animals , Blotting, Western , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/metabolism , Receptors, Somatotropin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...