Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Mol Nutr Food Res ; 66(23): e2200439, 2022 12.
Article in English | MEDLINE | ID: mdl-36153842

ABSTRACT

SCOPE: Fighting obesity and associated comorbidities through dieting is not always sustained and results in a subsequent weight gain, a phenomenon referred to as weight cycling. Diet is among the most important factors in modifying the composition of gut microbiota. The objective of this work is to determine whether weight cycling affects the composition and the predicted function of mouse fecal bacteria on a long-term basis. METHODS AND RESULTS: Mice fed for 40 weeks with either high fat (HF), low fat (LF), or cycled diets (starting and ending by one of the two diets, and the reverse) exhibit a bacterial profile specific to each of the four groups. A higher proportion of Firmicutes and Bacteroidota phyla are observed in mice on Hf and LF diet, respectively. The proportion of functions dedicated to amino acid metabolism is higher in mice on HF or LF/HF diets, whereas the mice on LF or HF/LF diets have a higher proportion of functions involve in carbohydrate metabolism and vitamin B biosynthesis. CONCLUSION: Compared to continuous HF or LF diets, cyclic diet specifically alters the composition and function of the mouse fecal microbiota, suggesting that fight against weight gain should be considered on a long-term basis.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Mice , Animals , Weight Cycling , Diet, High-Fat/adverse effects , Weight Gain , Bacteria , Mice, Inbred C57BL
2.
Endocrinology ; 163(7)2022 07 01.
Article in English | MEDLINE | ID: mdl-35617141

ABSTRACT

The gut microbiome has an important role in host development, metabolism, growth, and aging. Recent research points toward potential crosstalk between the gut microbiota and the growth hormone (GH)/insulin-like growth factor-1 (IGF-1) axis. Our laboratory previously showed that GH excess and deficiency are associated with an altered gut microbial composition in adult mice. Yet, no study to date has examined the influence of GH on the gut microbiome over time. Our study thus tracked the effect of excess GH action on the longitudinal changes in the gut microbial profile (ie, abundance, diversity/maturity, predictive metabolic function, and short-chain fatty acid [SCFA] levels) of bovine GH (bGH) transgenic mice at age 3, 6, and 12 months compared to littermate controls in the context of metabolism, intestinal phenotype, and premature aging. The bGH mice displayed age-dependent changes in microbial abundance, richness, and evenness. Microbial maturity was significantly explained by genotype and age. Moreover, several bacteria (ie, Lactobacillus, Lachnospiraceae, Bifidobacterium, and Faecalibaculum), predictive metabolic pathways (such as SCFA, vitamin B12, folate, menaquinol, peptidoglycan, and heme B biosynthesis), and SCFA levels (acetate, butyrate, lactate, and propionate) were consistently altered across all 3 time points, differentiating the longitudinal bGH microbiome from controls. Of note, the bGH mice also had significantly impaired intestinal fat absorption with increased fecal output. Collectively, these findings suggest that excess GH alters the gut microbiome in an age-dependent manner with distinct longitudinal microbial and predicted metabolic pathway signatures.


Subject(s)
Gastrointestinal Microbiome , Human Growth Hormone , Animals , Cattle , Fatty Acids, Volatile , Gastrointestinal Microbiome/genetics , Growth Hormone/metabolism , Male , Mice , Mice, Transgenic
3.
Pituitary ; 25(1): 116-130, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34373994

ABSTRACT

PURPOSE: Growth hormone (GH) has an important role in intestinal barrier function, and abnormalities in GH action have been associated with intestinal complications. Yet, the impact of altered GH on intestinal gross anatomy and morphology remains unclear. METHODS: This study investigated the influence of GH signaling on gross anatomy, morphology, and fibrosis by characterizing the small and large intestines in male and female bovine growth hormone transgenic (bGH) mice and GH receptor gene-disrupted (GHR-/-) mice at multiple timepoints. RESULTS: The length, weight, and circumference of the small and large intestines were increased in bGH mice and decreased in GHR-/- mice across all ages. Colon circumference was significantly increased in bGH mice in a sex-dependent manner while significantly decreased in male GHR-/- mice. Villus height, crypt depth, and muscle thickness of the small intestine were generally increased in bGH mice and decreased in GHR-/- mice compared to controls with age- and sex-dependent exceptions. Colonic crypt depth and muscle thickness in bGH and GHR-/- mice were significantly altered in an age- and sex-dependent manner. Fibrosis was increased in the small intestine of bGH males at 4 months of age, but no significant differences were seen between genotypes at other timepoints. CONCLUSION: This study observed notable opposing findings in the intestinal phenotype between mouse lines with GH action positively associated with intestinal gross anatomy (i.e. length, weight, and circumference). Moreover, GH action appears to alter morphology of the small and large intestines in an age- and sex-dependent manner.


Subject(s)
Growth Hormone , Intestine, Large/anatomy & histology , Intestine, Small/anatomy & histology , Receptors, Somatotropin , Age Factors , Animals , Cattle , Female , Male , Mice , Mice, Knockout , Receptors, Somatotropin/genetics , Sex Factors , Signal Transduction
4.
Pituitary ; 25(1): 1-51, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34797529

ABSTRACT

Much of our understanding of GH's action stems from animal models and the generation and characterization of genetically altered or modified mice. Manipulation of genes in the GH/IGF1 family in animals started in 1982 when the first GH transgenic mice were produced. Since then, multiple laboratories have altered mouse DNA to globally disrupt Gh, Ghr, and other genes upstream or downstream of GH or its receptor. The ability to stay current with the various genetically manipulated mouse lines within the realm of GH/IGF1 research has been daunting. As such, this review attempts to consolidate and summarize the literature related to the initial characterization of many of the known gene-manipulated mice relating to the actions of GH, PRL and IGF1. We have organized the mouse lines by modifications made to constituents of the GH/IGF1 family either upstream or downstream of GHR or to the GHR itself. Available data on the effect of altered gene expression on growth, GH/IGF1 levels, body composition, reproduction, diabetes, metabolism, cancer, and aging are summarized. For the ease of finding this information, key words are highlighted in bold throughout the main text for each mouse line and this information is summarized in Tables 1, 2, 3 and 4. Most importantly, the collective data derived from and reported for these mice have enhanced our understanding of GH action.


Subject(s)
Growth Hormone , Receptors, Somatotropin , Animals , Body Composition , Growth Hormone/genetics , Growth Hormone/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Transgenic , Models, Animal , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism
5.
Rev Endocr Metab Disord ; 22(1): 3-16, 2021 03.
Article in English | MEDLINE | ID: mdl-33033978

ABSTRACT

Nearly one century of research using growth hormone deficient (GHD) mouse lines has contributed greatly toward our knowledge of growth hormone (GH), a pituitary-derived hormone that binds and signals through the GH receptor and affects many metabolic processes throughout life. Although delayed sexual maturation, decreased fertility, reduced muscle mass, increased adiposity, small body size, and glucose intolerance appear to be among the negative characteristics of these GHD mouse lines, these mice still consistently outlive their normal sized littermates. Furthermore, the absence of GH action in these mouse lines leads to enhanced insulin sensitivity (likely due to the lack of GH's diabetogenic actions), delayed onset for a number of age-associated physiological declines (including cognition, cancer, and neuromusculoskeletal frailty), reduced cellular senescence, and ultimately, extended lifespan. In this review, we provide details about history, availability, growth, physiology, and aging of five commonly used GHD mouse lines.


Subject(s)
Disease Models, Animal , Growth Hormone/deficiency , Aging , Animals , Humans , Insulin Resistance , Mice , Obesity
6.
J Neuroendocrinol ; 32(11): e12893, 2020 11.
Article in English | MEDLINE | ID: mdl-33043505

ABSTRACT

Bovine growth hormone (bGH) transgenic mice mimic the clinical condition of acromegaly, having high circulating growth hormone (GH) levels. These mice are giant, have decreased adipose tissue (AT) mass, impaired glucose metabolism and a shortened lifespan. The detrimental effects of excess GH have been suggested, in part, to be a result of its depot-specific actions on AT. To investigate this relationship, we evaluated gene expression, biological mechanisms, cellular pathways and predicted microRNA (miRNA) in two AT depots (subcutaneous [Subq] and epididymal [Epi]) from bGH and littermate controls using RNA sequencing analysis. Two analyses on the differentially expressed genes (DEG) were performed: (i) comparison of the same AT depot between bGH and wild-type (WT) mice (genotype comparison) and (ii) comparison of Subq and Epi AT depots within the same genotype (depot comparison). For the genotype comparison, we found a higher number of significant DEG in the Subq AT depot of bGH mice compared to WT controls, corroborating previous reports that GH has a greater impact on the Subq depot. Furthermore, most of the DEG in bGH mice were not shared by WT mice, suggesting that excess GH induces the expression of genes not commonly present in AT. Through gene ontology and pathway analysis, the genotype comparison revealed that the DEG of the Subq depot of bGH mice relate to fatty acid oxidation, branched-chain amino acid degradation and the immune system. Additionally, the AT depot comparison showed that the immune cell activation and T-cell response appear up-regulated in the Subq compared to the Epi AT depot. The miRNA prediction also suggested a modulation of T-cell-related biological process in Subq. In summary, the present study provides a unique resource for understanding the specific differences in gene expression that are driven by both excess GH action and AT depot location.


Subject(s)
Adipose Tissue/metabolism , Growth Hormone/genetics , Growth Hormone/metabolism , Amino Acids, Branched-Chain/metabolism , Animals , Cattle , Epididymis/metabolism , Fatty Acids/metabolism , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Genotype , Immune System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/biosynthesis , MicroRNAs/genetics , Oxidation-Reduction , Signal Transduction/genetics , Subcutaneous Fat/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Growth Horm IGF Res ; 53-54: 101333, 2020.
Article in English | MEDLINE | ID: mdl-32717585

ABSTRACT

Both the GH/IGF-1 axis and the gut microbiota independently play an important role in host growth, metabolism, and intestinal homeostasis. Inversely, abnormalities in GH action and microbial dysbiosis (or a lack of diversity) in the gut have been implicated in restricted growth, metabolic disorders (such as chronic undernutrition, anorexia nervosa, obesity, and diabetes), and intestinal dysfunction (such as pediatric Crohn's disease, colonic polyps, and colon cancer). Over the last decade, studies have demonstrated that the microbial impact on growth may be mediated through the GH/IGF-1 axis, pointing toward a potential relationship between GH and the gut microbiota. This review covers current research on the GH/IGF-1 axis and the gut microbiome and its influence on overall host growth, metabolism, and intestinal health, proposing a bidirectional relationship between GH and the gut microbiome.


Subject(s)
Gastrointestinal Microbiome , Human Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Animals , Humans
8.
Endocrinology ; 161(8)2020 08 01.
Article in English | MEDLINE | ID: mdl-32556100

ABSTRACT

A rare 20K isoform of GH-V (here abbreviated as GHv) was discovered in 1998. To date, only 1 research article has characterized this isoform in vivo, observing that GHv treatment in male high-fat fed rats had several GH-like activities, but unlike GH lacked diabetogenic and lactogenic activities and failed to increase IGF-1 or body length. Therefore, the current study was conducted to further characterize the in vivo activities of GHv in a separate species and in a GH-deficient model (GH-/- mice) and with both sexes represented. GHv-treated GH-/- mice had significant increases to serum IGF-1, femur length, body length, body weight, and lean body mass and reduced body fat mass similar to mice receiving GH treatment. GH treatment increased circulating insulin levels and impaired insulin sensitivity; in contrast, both measures were unchanged in GHv-treated mice. Since GHv lacks prolactin receptor (PRLR) binding activity, we tested the ability of GH and GHv to stimulate the proliferation of human cancer cell lines and found that GHv has a decreased proliferative response in cancers with high PRLR. Our findings demonstrate that GHv can stimulate insulin-like growth factor-1 and subsequent longitudinal body growth in GH-deficient mice similar to GH, but unlike GH, GHv promoted growth without inhibiting insulin action and without promoting the growth of PRLR-positive cancers in vitro. Thus, GHv may represent improvements to current GH therapies especially for individuals at risk for metabolic syndrome or PRLR-positive cancers.


Subject(s)
Growth Hormone/genetics , Human Growth Hormone/pharmacology , Placental Hormones/pharmacology , Animals , Body Composition/drug effects , Body Weight/drug effects , Female , Growth Hormone/deficiency , Hormone Replacement Therapy , Human Growth Hormone/isolation & purification , Human Growth Hormone/metabolism , Human Growth Hormone/therapeutic use , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Placenta/chemistry , Placenta/metabolism , Placental Hormones/therapeutic use , Pregnancy , Protein Isoforms
9.
Endocrinology ; 161(4)2020 04 01.
Article in English | MEDLINE | ID: mdl-32100023

ABSTRACT

The gut microbiome has been implicated in host metabolism, endocrinology, and pathophysiology. Furthermore, several studies have shown that gut bacteria impact host growth, partially mediated through the growth hormone (GH)/insulin-like growth factor 1 (IGF-1) axis. Yet, no study to date has examined the specific role of GH on the gut microbiome. Our study thus characterized the adult gut microbial profile and intestinal phenotype in GH gene-disrupted (GH-/-) mice (a model of GH deficiency) and bovine GH transgenic (bGH) mice (a model of chronic, excess GH action) at 6 months of age. Both the GH-/- and bGH mice had altered microbial signatures, in opposing directions at the phylum and genus levels. For example, GH-/- mice had significantly reduced abundance in the Proteobacteria, Campylobacterota, and Actinobacteria phyla, whereas bGH mice exhibited a trending increase in those phyla compared with respective controls. Analysis of maturity of the microbial community demonstrated that lack of GH results in a significantly more immature microbiome while excess GH increases microbial maturity. Several common bacterial genera were shared, although in opposing directions, between the 2 mouse lines (e.g., decreased in GH-/- mice and increased in bGH mice), suggesting an association with GH. Similarly, metabolic pathways like acetate, butyrate, heme B, and folate biosynthesis were predicted to be impacted by GH. This study is the first to characterize the gut microbiome in mouse lines with altered GH action and indicates that GH may play a role in the growth of certain microbiota thus impacting microbial maturation and metabolic function.


Subject(s)
Dwarfism, Pituitary/microbiology , Gastrointestinal Microbiome/physiology , Growth Hormone/metabolism , Animals , Dwarfism, Pituitary/genetics , Dwarfism, Pituitary/metabolism , Growth Hormone/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic
10.
Arch. endocrinol. metab. (Online) ; 63(6): 557-567, Nov.-Dec. 2019. tab, graf
Article in English | LILACS | ID: biblio-1055021

ABSTRACT

ABSTRACT In order to provide new insights into the various activities of GH in specific tissues, recent advances have allowed for the generation of tissue-specific GHR knockout mice. To date, 21 distinct tissue-specific mouse lines have been created and reported in 28 publications. Targeted tissues include liver, muscle, fat, brain, bone, heart, intestine, macrophage, pancreatic beta cells, hematopoietic stem cells, and multi-tissue "global". In this review, we provide a brief history and description of the 21 tissue-specific GHR knockout mouse lines. Arch Endocrinol Metab. 2019;63(6):557-67


Subject(s)
Animals , Rats , Receptors, Somatotropin/physiology , Growth Hormone/physiology , Signal Transduction , Mice, Knockout , Models, Animal
11.
Biotechniques ; 67(2): 55-62, 2019 08.
Article in English | MEDLINE | ID: mdl-31290335

ABSTRACT

Illumina-based amplicon sequencing suffers from the deleterious effects of highly homogenous nucleotide composition, limiting the number of high-quality reads generated per run. We attempted to alleviate this limitation by comparing the results obtained from 16S ribosomal DNA (16S rDNA) sequencing of mouse gut microbiomes using Illumina V3-V4 primers (Run 1) and custom primers that incorporate a heterogeneity spacer (0-7 nucleotides) upstream of the 16S priming region (Run 2). Overall, Run 2 had higher quality sequences, a more diverse microbial profile, and higher precision within, and variation between, experimental groups than Run 1. Our primer design offers a simple way to increase the quality of 16S rDNA sequencing and increases the number of useable reads generated per Illumina run.


Subject(s)
Feces/microbiology , Gastrointestinal Microbiome , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA/methods , Animals , DNA Primers/genetics , Female , Gene Library , Mice , Mice, Inbred C57BL
13.
Endocrinology ; 160(7): 1743-1756, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31099824

ABSTRACT

In 1997, our laboratory used targeted gene disruption of the GH receptor (GHR) to generate GHR knockout (GHR-/-) mice, which have been used in >127 published studies to help elucidate GH's numerous activities. However, because GH replacement studies cannot be performed using this line, a GH knockout mouse line via targeted disruption of the GH gene is needed. Therefore, we created and characterized GH gene-disrupted (GH-/-) mice. GH-/- mice have severely decreased IGF-1 levels, small body size, and altered body composition with increased adiposity. GH-/- mice are extremely insulin sensitive but glucose intolerant, with a dramatic reduction in pancreatic islet size. Importantly, disruption of the GH gene had profound and depot-specific effects on white adipose tissue (WAT). Subcutaneous WAT from male and female GH-/- mice have significantly larger adipocytes and reduced fibrosis, neither of which occurred in perigonadal WAT, suggesting that GH has a more pronounced effect on subcutaneous WAT. Comparisons of GH-/- mice to previously published data on GHR-/- mice show a remarkably similar phenotype. Finally, we demonstrate that GH-/- mice are responsive to GH treatment, as shown by changes to serum IGF-1 levels; body length, weight, and composition; and insulin sensitivity. This study not only provides characterization of the first mouse line with targeted mutation of the GH gene but also indicates that GH gene disruption dramatically influences fibrosis of subcutaneous WAT.


Subject(s)
Adipocytes/metabolism , Growth Hormone/genetics , Insulin Resistance/physiology , Subcutaneous Fat/metabolism , Adipose Tissue, White/metabolism , Animals , Body Composition/physiology , Female , Fibrosis/genetics , Fibrosis/metabolism , Growth Hormone/metabolism , Islets of Langerhans/metabolism , Male , Mice , Mice, Knockout
14.
Growth Horm IGF Res ; 46-47: 5-15, 2019.
Article in English | MEDLINE | ID: mdl-31078722

ABSTRACT

OBJECTIVE: Growth hormone (GH) has been reported to enhance the intestinal barrier; as such, recombinant GH has been administered for several intestinal diseases. However, excess GH action has been implicated in increasing the risk of intestinal dysfunction. The goal of this study was to examine the direct effects of GH on the small and large intestines to clarify the role GH plays in intestinal function through the use of a mouse model. DESIGN: An intestinal epithelial-specific GH receptor (GHR) knockout (IntGHRKO) mouse line was generated using Cre-lox with the villin promoter driving Cre expression. The generated mice were characterized with respect to growth and intestinal phenotypes. RESULTS: IntGHRKO mice showed no significant changes in body length, weight, or composition compared to floxed controls. Male IntGHRKO mice had significantly shorter large intestines at 4 and 12 months of age. Intestinal barrier function was assessed by measuring the expression of tight junction related genes, as well as levels of serum endotoxin and fecal albumin. Results showed sex differences as males had an increase in occludin levels but normal serum endotoxin and fecal albumin; while, females had changes in fecal albumin levels with normal occludin and serum endotoxin. Evaluation of glucose tolerance and fat absorption also showed sex differences as females were glucose intolerant, while males had impaired fat absorption. Histopathology revealed a trend towards decreased villus height in males, which could explain the sex difference in glucose homeostasis. CONCLUSIONS: Overall, the data demonstrate that disruption of GH on the intestinal epithelial cells modestly affects the intestinal gross anatomy, morphology, and function in a sex-specific manner.


Subject(s)
Glucose/metabolism , Homeostasis , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Intestinal Mucosa/metabolism , Receptors, Somatotropin/physiology , Animals , Body Weight , Female , Gene Knockout Techniques , Integrases/metabolism , Male , Mice , Mice, Knockout
15.
Arch Endocrinol Metab ; 63(6): 557-567, 2019.
Article in English | MEDLINE | ID: mdl-31939480

ABSTRACT

In order to provide new insights into the various activities of GH in specific tissues, recent advances have allowed for the generation of tissue-specific GHR knockout mice. To date, 21 distinct tissue-specific mouse lines have been created and reported in 28 publications. Targeted tissues include liver, muscle, fat, brain, bone, heart, intestine, macrophage, pancreatic beta cells, hematopoietic stem cells, and multi-tissue "global". In this review, we provide a brief history and description of the 21 tissue-specific GHR knockout mouse lines. Arch Endocrinol Metab. 2019;63(6):557-67.


Subject(s)
Growth Hormone/physiology , Receptors, Somatotropin/physiology , Animals , Mice , Mice, Knockout , Models, Animal , Signal Transduction
16.
Endocrinology ; 160(1): 68-80, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30462209

ABSTRACT

Global GH receptor-null or knockout (GHRKO) mice have been extensively studied owing to their unique phenotype (dwarf and obese but remarkably insulin sensitive and long-lived). To better understand the influence of adipose tissue (AT) on the GHRKO phenotype, we previously generated fat-specific GHRKO (FaGHRKO) mice using the adipocyte protein-2 (aP2) promoter driving Cre expression. Unlike global GHRKO mice, FaGHRKO mice are larger than control mice and have an increase in white AT (WAT) mass and adipocyte size as well as an increase in brown AT mass. FaGHRKO mice also have an unexpected increase in IGF-1, decrease in adiponectin, no change in insulin sensitivity or liver triglyceride content, and a decreased lifespan. Extensive analysis of the aP2 promoter/enhancer by multiple laboratories has revealed expression in nonadipose tissues, confounding interpretation of results. In the current study, we used the adiponectin promoter/enhancer to drive Cre expression, which better targets mature adipocytes, and generated a new line of adipocyte-specific GHRKO (AdGHRKO) mice. AdGHRKO mice have an increase in adipocyte size and WAT depot mass in all depots except male perigonadal, a WAT accumulation pattern similar to FaGHRKO mice. Likewise, adiponectin levels and WAT fibrosis are decreased in both tissue-specific mouse lines. However, unlike FaGHRKO mice, AdGHRKO mice have no change in IGF-1 levels, improved glucose homeostasis, and reduced liver triglycerides. Thus, AdGHRKO mice should be valuable for future studies assessing the contribution of adipocyte GHR signaling in long-term health and lifespan.


Subject(s)
Adipocytes/metabolism , Carrier Proteins/genetics , Insulin Resistance , Liver/metabolism , Triglycerides/metabolism , Adiponectin , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Carrier Proteins/metabolism , Female , Humans , Insulin/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Knockout , Species Specificity
17.
Analyst ; 142(20): 3797-3799, 2017 Oct 09.
Article in English | MEDLINE | ID: mdl-28920605

ABSTRACT

We discuss and demonstrate why FTIR/ATR spectra can only be calibrated in wavelength, not intensity, for comparison with other data sets at present. This is because the intensity calibration must remove the instrument response function. To address this problem, we suggest a possible approach.

18.
Compr Physiol ; 7(3): 819-840, 2017 06 18.
Article in English | MEDLINE | ID: mdl-28640444

ABSTRACT

Increasing prevalence of obesity and obesity-related conditions worldwide has necessitated a more thorough understanding of adipose tissue (AT) and expanded the scope of research in this field. AT is now understood to be far more complex and dynamic than previously thought, which has also fueled research to reevaluate how hormones, such as growth hormone (GH), alter the tissue. In this review, we will introduce properties of AT important for understanding how GH alters the tissue, such as anatomical location of depots and adipokine output. We will provide an overview of GH structure and function and define several human conditions and cognate mouse lines with extremes in GH action that have helped shape our understanding of GH and AT. A detailed discussion of the GH/AT relationship will be included that addresses adipokine production, immune cell populations, lipid metabolism, senescence, differentiation, and fibrosis, as well as brown AT and beiging of white AT. A brief overview of how GH levels are altered in an obese state, and the efficacy of GH as a therapeutic option to manage obesity will be given. As we will reveal, the effects of GH on AT are numerous, dynamic and depot-dependent. © 2017 American Physiological Society. Compr Physiol 7:819-840, 2017.


Subject(s)
Adipose Tissue/metabolism , Growth Hormone/metabolism , Adipose Tissue/pathology , Animals , Growth Hormone/genetics , Humans , Obesity/metabolism , Obesity/pathology , Pituitary Diseases/metabolism , Pituitary Diseases/pathology , Signal Transduction
19.
Nurse Pract ; 40(4): 34-9, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25774813

ABSTRACT

Early recognition of developmental delay is critical to providing comprehensive pediatric primary care. Advanced practice nurses must be aware of the guidelines for surveillance and developmental screening in children. This article discusses guidelines for screening, examples of screening tools, information for follow up, and referral for positive screenings.


Subject(s)
Developmental Disabilities/nursing , Mass Screening/nursing , Population Surveillance/methods , Practice Guidelines as Topic , Primary Care Nursing/methods , Advanced Practice Nursing , Child , Child, Preschool , Early Diagnosis , Humans , Infant , Infant, Newborn , Mass Screening/methods , Nursing Diagnosis , Nursing Methodology Research , Pediatric Nurse Practitioners
20.
Endocrinology ; 155(5): 1793-805, 2014 May.
Article in English | MEDLINE | ID: mdl-24517230

ABSTRACT

GH is an important regulator of body growth and composition as well as numerous other metabolic processes. In particular, liver plays a key role in the GH/IGF-I axis, because the majority of circulating "endocrine" IGF-I results from GH-stimulated liver IGF-I production. To develop a better understanding of the role of liver in the overall function of GH, we generated a strain of mice with liver-specific GH receptor (GHR) gene knockout (LiGHRKO mice). LiGHRKO mice had a 90% decrease in circulating IGF-I levels, a 300% increase in circulating GH, and significant changes in IGF binding protein (IGFBP)-1, IGFBP-2, IGFBP-3, IGFBP-5, and IGFBP-7. LiGHRKO mice were smaller than controls, with body length and body weight being significantly decreased in both sexes. Analysis of body composition over time revealed a pattern similar to those found in GH transgenic mice; that is, LiGHRKO mice had a higher percentage of body fat at early ages followed by lower percentage of body fat in adulthood. Local IGF-I mRNA levels were significantly increased in skeletal muscle and select adipose tissue depots. Grip strength was increased in LiGHRKO mice. Finally, circulating levels of leptin, resistin, and adiponectin were increased in LiGHRKO mice. In conclusion, LiGHRKO mice are smaller despite increased local mRNA expression of IGF-I in several tissues, suggesting that liver-derived IGF-I is indeed important for normal body growth. Furthermore, our data suggest that novel GH-dependent cross talk between liver and adipose is important for regulation of adipokines in vivo.


Subject(s)
Adipokines/metabolism , Aging , Endocrine Glands/metabolism , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Liver/metabolism , Receptors, Somatotropin/metabolism , Adipose Tissue, White/growth & development , Adipose Tissue, White/metabolism , Adiposity , Animals , Body Composition , Body Size , Female , Growth Hormone/blood , Insulin-Like Growth Factor I/genetics , Liver/growth & development , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle Development , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , RNA, Messenger/metabolism , Receptors, Somatotropin/genetics , Sex Characteristics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...