Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Aging Cell ; : e14226, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38808779

ABSTRACT

Dysregulation of growth hormone (GH) signaling consistently leads to increased lifespan in laboratory rodents, yet the precise mechanisms driving this extension remain unclear. Understanding the molecular underpinnings of the beneficial effects associated with GH deficiency could unveil novel therapeutic targets for promoting healthy aging and longevity. In our pursuit of identifying metabolites implicated in aging, we conducted an unbiased lipidomic analysis of serum samples from growth hormone-releasing hormone knockout (GHRH-KO) female mice and their littermate controls. Employing a targeted lipidomic approach, we specifically investigated ceramide levels in GHRH-KO mice, a well-established model of enhanced longevity. While younger GHRH-KO mice did not exhibit notable differences in serum lipids, older counterparts demonstrated significant reductions in over one-third of the evaluated lipids. In employing the same analysis in liver tissue, GHRH-KO mice showed pronounced downregulation of numerous ceramides and hexosylceramides, which have been shown to elicit many of the tissue defects that accompany aging (e.g., insulin resistance, oxidative stress, and cell death). Additionally, gene expression analysis in the liver tissue of adult GHRH-KO mice identified substantial decreases in several ceramide synthesis genes, indicating that these alterations are, at least in part, attributed to GHRH-KO-induced transcriptional changes. These findings provide the first evidence of disrupted ceramide metabolism in a long-lived mammal. This study sheds light on the intricate connections between GH deficiency, ceramide levels, and the molecular mechanisms influencing lifespan extension.

2.
Aging Cell ; 23(3): e14051, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38279509

ABSTRACT

The Methionine restriction (MR) diet has been shown to delay aging and extend lifespan in various model organisms. However, the long-term effects of MR diet on the gut microbiome composition remain unclear. To study this, male mice were started on MR and control diet regimens at 6 months and continued until 22 months of age. MR mice have reduced body weight, fat mass percentage, and bone mineral density while having increased lean mass percentage. MR mice also have increased insulin sensitivity along with increasing indirect calorimetry markers such as energy expenditure, oxygen consumption, carbon dioxide production, and glucose oxidation. Fecal samples were collected at 1 week, 18 weeks, and 57 weeks after the diet onset for 16S rRNA amplicon sequencing to study the gut microbiome composition. Alpha and beta diversity metrics detected changes occurring due to the timepoint variable, but no changes were detected due to the diet variable. The results from LEfSe analysis surprisingly showed that more bacterial taxa changes were linked to age rather than diet. Interestingly, we found that the long-term MR diet feeding induced smaller changes compared to short-term feeding. Specific taxa changes due to the diet were observed at the 1 or 18-week time points, including Ileibacterium, Odoribacter, Lachnoclostridium, Marinifilaceae, and Lactobacillaceae. Furthermore, there were consistent aging-associated changes across both groups, with an increase in Ileibacterium and Erysipelotrichaceae with age, while Eubacterium_coprostanoligenes_group, Ruminococcaceae, Peptococcaceae, and Peptococcus decreased with age.


Subject(s)
Gastrointestinal Microbiome , Methionine , Male , Mice , Animals , Gastrointestinal Microbiome/genetics , RNA, Ribosomal, 16S , Diet , Body Weight , Racemethionine/pharmacology
3.
Aging Cell ; 22(12): e13985, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37667562

ABSTRACT

Our previous research has demonstrated that mice lacking functional growth hormone-releasing hormone (GHRH) exhibit distinct physiological characteristics, including an extended lifespan, a preference for lipid utilization during rest, mild hypoglycemia, and heightened insulin sensitivity. They also show a further increase in lifespan when subjected to caloric restriction. These findings suggest a unique response to fasting, which motivated our current study on the response to glucagon, a key hormone released from the pancreas during fasting that regulates glucose levels, energy expenditure, and metabolism. Our study investigated the effects of an acute glucagon challenge on female GHRH knockout mice and revealed that they exhibit reduced glucose production, likely due to suppressed gluconeogenesis. However, these mice showed an increase in energy expenditure. We also observed alterations in pancreatic islet architecture, with smaller islets and a reduction of insulin-producing beta cells but no changes in glucagon-producing alpha cells. Additionally, the analysis of hepatic glucagon signaling showed a decrease in glucagon receptor expression and phosphorylated CREB. In conclusion, our findings suggest that the unique metabolic phenotype observed in these long-lived mice may be partly explained by changes in glucagon signaling. Further exploration of this pathway may lead to new insights into the regulation of longevity in mammals.


Subject(s)
Glucagon , Longevity , Female , Mice , Animals , Glucagon/metabolism , Glucagon/pharmacology , Mice, Knockout , Longevity/genetics , Insulin/metabolism , Growth Hormone-Releasing Hormone , Glucose/metabolism , Mammals/metabolism
4.
Biomedicines ; 11(8)2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37626614

ABSTRACT

L-serine is a non-essential amino acid that plays a vital role in protein synthesis, cell proliferation, development, and sphingolipid formation in the central nervous system. It exerts its effects through the activation of glycine receptors and upregulation of PPAR-γ, resulting in neurotransmitter synthesis, neuroprotection, and anti-inflammatory effects. L-serine shows potential as a protective agent in various neurological diseases and neurodegenerative disorders. Deficiency of L-serine and its downstream products has been linked to severe neurological deficits. Despite its crucial role, there is limited understanding of its mechanistic production and impact on glial and neuronal cells. Most of the focus has been on D-serine, the downstream product of L-serine, which has been implicated in a wide range of neurological diseases. However, L-serine is approved by FDA for supplemental use, while D-serine is not. Hence, it is imperative that we investigate the wider effects of L-serine, particularly in relation to the pathogenesis of several neurological deficits that, in turn, lead to diseases. This review aims to explore current knowledge surrounding L-serine and its potential as a treatment for various neurological diseases and neurodegenerative disorders.

5.
Aging Cell ; 22(7): e13854, 2023 07.
Article in English | MEDLINE | ID: mdl-37095621

ABSTRACT

Alzheimer's disease (AD), a prevalent form of dementia, is characterized by the decline of cognitive abilities with age. Available treatment options for AD are limited, making it a significant public health concern. Recent research suggests that metabolic dysfunction plays a role in the development of AD. In addition, insulin therapy has been shown to improve memory in patients with cognitive decline. In this study, we report the first examination of body composition, peripheral insulin sensitivity, and glucose tolerance in relation to behavioral assessments of learning, memory, and anxiety in the TgF344-AD rat model of AD. Results from glucose and insulin tolerance tests show that female TgF344-AD rats exhibit impaired glucose clearance and reduced insulin sensitivity at both 9 and 12 months of age, while males display no differences at 9 months and even improved glucose clearance at 12 months. Results from the Morris Water Maze assessment of learning and memory reveal that male TgF344-AD rats display impairments at both 9 and 12 months of age, while female TgF344-AD rats only show impairments at 12 months. Furthermore, results from open field and elevated plus maze tests suggest that female TgF344-AD rats display increased anxiety at 9 months of age; however, no differences were detected in males or at 12 months of age. Overall, our findings suggest that impairments in metabolism, commonly associated with type 2 diabetes, occur before or simultaneously with cognitive decline and anxiety in a sexually dimorphic manner in the TgF344-AD rat model.


Subject(s)
Alzheimer Disease , Diabetes Mellitus, Type 2 , Insulin Resistance , Rats , Male , Female , Animals , Alzheimer Disease/metabolism , Rats, Transgenic , Rats, Inbred F344 , Sex Characteristics , Diabetes Mellitus, Type 2/complications , Insulin , Homeostasis , Glucose , Disease Models, Animal
6.
Aging (Albany NY) ; 15(2): 459-471, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36640271

ABSTRACT

Alzheimer's disease (AD) is one of the most devastating diseases currently in the world with no effective treatments. There is increasing evidence that the gut microbiome plays a role in AD. Here we set out to study the age-related changes in the microbiome of the Tgf344-AD rats. We performed 16S ribosomal RNA sequencing on the fecal samples of male rats at 14 and 20 months of age. We found the Tgf344-AD rats to have decreased microbial diversity compared to controls at 14 months of age and this was found to be opposite at 20 months of age. Interestingly, we found a distinctive shift in the microbial community structure of the rats with aging along with changes in the microbiota composition. Some of the observed changes in the Tgf344AD rats were in the genera Bifidobacterium, Ruminococcus, Parasutterella, Lachnoclostridium and Butyricicoccus. Other age-related changes occuring in both the Tgf344-AD and WT control rats were decreases in Enterohaldus, Escherichia Shigella, Rothia and increase in Turicibacter and Clostrium_senso_stricto. Our study has shown that gut microbiota changes occurs in this Alzheimer's disease rat model.


Subject(s)
Alzheimer Disease , Gastrointestinal Microbiome , Microbiota , Rats , Male , Animals , Alzheimer Disease/genetics , Gastrointestinal Microbiome/genetics , Aging , Feces/microbiology
7.
Biomedicines ; 10(8)2022 Aug 06.
Article in English | MEDLINE | ID: mdl-36009454

ABSTRACT

While glucagon has long been recognized as the primary counter hormone to insulin's actions, it has recently gained recognition as a metabolic regulator with its effects extending beyond control of glycemia. Recently developed models of tissue-specific glucagon receptor knockouts have advanced our understanding of this hormone, providing novel insight into the role it plays within organs as well as its systemic effects. Studies where the pharmacological blockade of the glucagon receptor has been employed have proved similarly valuable in the study of organ-specific and systemic roles of glucagon signaling. Studies carried out employing these tools demonstrate that glucagon indeed plays a role in regulating glycemia, but also in amino acid and lipid metabolism, systemic endocrine, and paracrine function, and in the response to cardiovascular injury. Here, we briefly review recent progress in our understanding of glucagon's role made through inhibition of glucagon receptor signaling utilizing glucagon receptor antagonists and tissue specific genetic knockout models.

8.
Front Endocrinol (Lausanne) ; 13: 797581, 2022.
Article in English | MEDLINE | ID: mdl-35282433

ABSTRACT

It is well documented that the environment of the developing fetus, including availability of nutrients and presence of toxins, can have major impact on adult phenotype, age-related traits and risk of chronic disease. There is also accumulating evidence that postnatal environment can impact adult characteristics related to evolutionary fitness, health, and aging. To determine whether early life hormonal interventions can alter trajectory of aging, we have examined the effects of early life growth hormone (GH) replacement therapy in Prop1df (Ames dwarf) mice which are GH deficient and remarkably long lived. Twice-daily GH injections between the ages of two and eight weeks completely normalized ("rescued") a number of adult metabolic characteristics believed to contribute to extended longevity of these mutants. Importantly, longevity of Ames dwarf mice was reduced by early life GH treatment. This was associated with histone H3 modifications. We conclude that the trajectory of mammalian aging can be modified by early life interventions. Mechanistic links among interventions during postnatal development, adult metabolic characteristics, aging, and longevity, apparently involve epigenetic phenomena.


Subject(s)
Dwarfism , Growth Hormone , Aging , Animals , Dwarfism/genetics , Dwarfism/metabolism , Growth Hormone/metabolism , Hormone Replacement Therapy , Longevity , Mammals/metabolism , Mice
9.
Biomedicines ; 11(1)2022 Dec 30.
Article in English | MEDLINE | ID: mdl-36672612

ABSTRACT

Neurodegenerative diseases feature changes in cognition, and anxiety-like and autism-like behaviors, which are associated with epigenetic alterations such as DNA methylation and histone modifications. The amino acid L-serine has been shown to have beneficial effects on neurological symptoms. Here, we found that growth hormone-releasing hormone knockout (GHRH-KO) mice, a GH-deficiency mouse model characterized by extended lifespan and enhanced insulin sensitivity, showed a lower anxiety symptom and impairment of short-term object recognition memory and autism-like behaviors. Interestingly, L-serine administration exerted anxiolytic effects in mice and ameliorated the behavioral deficits in GHRH-KO. L-serine treatment upregulated histone epigenetic markers of H3K4me, H3K9ac, H3K14ac and H3K18ac in the hippocampus and H3K4me in the cerebral cortex in both GHRH-KO mice and wild type controls. L-serine-modulated epigenetic marker changes, in turn, were found to regulate mRNA expression of BDNF, grm3, foxp1, shank3, auts2 and marcksl1, which are involved in anxiety-, cognitive- and autism-like behaviors. Our study provides a novel insight into the beneficial effects of L-serine intervention on neuropsychological impairments.

10.
Geroscience ; 43(5): 2149-2160, 2021 10.
Article in English | MEDLINE | ID: mdl-34304389

ABSTRACT

The UAB Nathan Shock Center focuses on comparative energetics and aging. Energetics, as defined for this purpose, encompasses the causes, mechanisms, and consequences of the acquisition, storage, and use of metabolizable energy. Comparative energetics is the study of metabolic processes at multiple scales and across multiple species as it relates to health and aging. The link between energetics and aging is increasingly understood in terms of dysregulated mitochondrial function, altered metabolic signaling, and aberrant nutrient responsiveness with increasing age. The center offers world-class expertise in comprehensive, integrated energetic assessment and analysis from the level of the organelle to the organism and across species from the size of worms to rats as well as state-of-the-art data analytics. The range of services offered by our three research cores, (1) The Organismal Energetics Core, (2) Mitometabolism Core, and (3) Data Analytics Core, is described herein.


Subject(s)
Aging , Mitochondria , Animals , Rats , Signal Transduction
11.
Aging Cell ; 20(4): e13339, 2021 04.
Article in English | MEDLINE | ID: mdl-33755309

ABSTRACT

Mice with disruptions of growth hormone-releasing hormone (GHRH) or growth hormone receptor (GHR) exhibit similar phenotypes of prolonged lifespan and delayed age-related diseases. However, these two models respond differently to calorie restriction indicating that they might carry different and/or independent mechanisms for improved longevity and healthspan. In order to elucidate these mechanisms, we generated GHRH and GHR double-knockout mice (D-KO). In the present study, we focused specifically on the characteristics of female D-KO mice. The D-KO mice have reduced body weight and enhanced insulin sensitivity compared to wild-type (WT) controls. Growth retardation in D-KO mice is accompanied by decreased GH expression in pituitary, decreased circulating IGF-1, increased high-molecular-weight (HMW) adiponectin, and leptin hormones compared to WT controls. Generalized linear model-based regression analysis, which controls for body weight differences between D-KO and WT groups, shows that D-KO mice have decreased lean mass, bone mineral density, and bone mineral content, but increased adiposity. Indirect calorimetry markers including oxygen consumption, carbon dioxide production, and energy expenditure were significantly lower in D-KO mice relative to the controls. In comparison with WT mice, the D-KO mice displayed reduced respiratory exchange ratio (RER) values only during the light cycle, suggesting a circadian-related metabolic shift toward fat utilization. Interestingly, to date survival data suggest extended lifespan in D-KO female mice.


Subject(s)
Aging/genetics , Aging/metabolism , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Longevity/genetics , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , Signal Transduction/genetics , Adiposity/genetics , Animals , Body Weight/genetics , Caloric Restriction , Carbon Dioxide/metabolism , Energy Metabolism/genetics , Female , Gene Editing/methods , Gene Knockout Techniques/methods , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Leptin/metabolism , Mice , Mice, Knockout , Oxygen Consumption/genetics
12.
Curr Dev Nutr ; 5(12): nzab134, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34993387

ABSTRACT

BACKGROUND: Poultry eggs are a low-cost, high-protein nutrient package that can be consumed as part of quality diets. However, consumption of poultry egg products is historically contentious, which highlights the importance of investigating impacts of long-term egg consumption on metabolic health. OBJECTIVE: Our study utilized the zebrafish, Danio rerio, a newly defined model of human metabolic health, to understand the metabolic consequence of consuming egg products in lieu of other well-described protein sources. METHODS: Reference diets were formulated to contain multisource protein with casein and fish protein hydrolysate (CON; control protein sources), the protein sources that have been historically utilized in numerous reference diets. These proteins were then partially replaced with either whole egg (WE; protein and lipid source), egg white (EW; protein source), wheat gluten (WG; cereal protein source), or a high-lipid-content diet containing a multisource protein with casein and fish protein hydrolysate (HFCON; isonitrogenous and isolipidic with the WE diet) in a 34-wk trial (n = 8 tanks, 10 fish per tank). Daily feeding was initiated at the early juvenile life stage and terminated at the late reproductive adult stage. RESULTS: The amino acid composition of control versus egg product diets did not vary substantially, although methionine and lysine were apparently limiting in fish fed WG. At termination, fish fed EW as the protein source had weight gain and body composition similar to those fed the CON diet. Fasting and postprandial blood glucose did not differ between any dietary treatment. Assessment of the liver transcriptome using RNAseq revealed no differential gene expression between zebrafish fed CON or WE diets. Zebrafish fed WG had lower weight gain in males. CONCLUSIONS: Long-term consumption of egg products promoted metabolic health equal to that of historically relevant proteins. These data support the value of egg products for maintaining long-term metabolic health in animal diets.

13.
Aging (Albany NY) ; 13(2): 1633-1648, 2020 12 28.
Article in English | MEDLINE | ID: mdl-33378746

ABSTRACT

Histone modifications, specifically in the lysine residues of histone H3, have been implicated in lifespan regulation in several model organisms. Our previous studies showed that growth hormone (GH) treatment during early life can dramatically influence lifespan in long-lived Ames dwarf mice. However, the effects of this hormonal intervention on epigenetic modifications have never been examined. In this study, we sought to compare tissue-specific histone H3 lysine methylation and acetylation markers in Ames dwarf and wild type (WT) mice and to determine how these markers are affected by early-life GH intervention. Ames dwarf mice exhibited suppressed H3K4me in both hepatic and brain tissues, while showing elevated H3K27me in the brain. Early-life GH intervention significantly altered the histone H3 markers in those tissues. Furthermore, early GH intervention increased expression of histone H3 acetylation at multiple lysine residues in a tissue-specific manner. This included changes in H3K14ac and H3K18ac in the liver and brain, H3K18ac in visceral adipose tissue and H3K9ac, H3K14ac and H3K27ac in subcutaneous adipose tissue. This study serves as an initial, but important step in elucidating the epigenetic mechanisms by which hormonal signals during early life can influence aging and longevity in mammals.


Subject(s)
Brain/drug effects , Dwarfism, Pituitary/metabolism , Epigenesis, Genetic/drug effects , Growth Hormone/pharmacology , Histones/drug effects , Liver/drug effects , Acetylation/drug effects , Animals , Brain/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/drug effects , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferases/drug effects , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Disease Models, Animal , Dwarfism, Pituitary/genetics , Enhancer of Zeste Homolog 2 Protein , Growth Hormone/deficiency , Histone Code/drug effects , Histones/metabolism , Homeodomain Proteins/genetics , Hormone Replacement Therapy , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Liver/metabolism , Longevity/genetics , Methylation/drug effects , Mice , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism , DNA Methyltransferase 3B
14.
Front Endocrinol (Lausanne) ; 11: 579909, 2020.
Article in English | MEDLINE | ID: mdl-33162937

ABSTRACT

Growth hormone (GH) signaling plays a key role in mediating growth, development, metabolism, and lifespan regulation. However, the mechanisms of longevity regulation at the cellular and molecular level are still not well-understood. An important area in the field of GH research is in the development of advanced transgenic systems for conditional expression of GH signaling in a cell type- or tissue-specific manner. There have been many recent studies conducted to examine the effects of tissue-specific GHR disruption. This review updates our previous discussions on this topic and summarizes recent data on the newly-made tissue-specific GHR-KO mice including intestinal epithelial cells, bone, hematopoietic stem cells, cardiac myocytes, and specific brain regions. The data from these new genetically-engineered mice have a significant impact on our understanding of the local GH signaling function.


Subject(s)
Growth Hormone/metabolism , Homeostasis , Receptors, Somatotropin/physiology , Animals , Longevity , Mice , Mice, Knockout , Organ Specificity
15.
Genes (Basel) ; 11(9)2020 09 18.
Article in English | MEDLINE | ID: mdl-32962067

ABSTRACT

Aging is a complex process mainly categorized by a decline in tissue, cells and organ function and an increased risk of mortality. Recent studies have provided evidence that suggests a strong association between epigenetic mechanisms throughout an organism's lifespan and age-related disease progression. Epigenetics is considered an evolving field and regulates the genetic code at several levels. Among these are DNA changes, which include modifications to DNA methylation state, histone changes, which include modifications of methylation, acetylation, ubiquitination and phosphorylation of histones, and non-coding RNA changes. As a result, these epigenetic modifications are vital targets for potential therapeutic interventions against age-related deterioration and disease progression. Dietary polyphenols play a key role in modulating these modifications thereby delaying aging and extending longevity. In this review, we summarize recent advancements linking epigenetics, polyphenols and aging as well as critical findings related to the various dietary polyphenols in different fruits and vegetables. In addition, we cover studies that relate polyphenols and their epigenetic effects to various aging-related diseases such as cardiovascular diseases, neurodegenerative diseases, autoimmune disorders, diabetes, osteoporosis and cancer.


Subject(s)
Aging/drug effects , Cardiovascular Diseases/prevention & control , DNA Methylation , Epigenesis, Genetic , Neoplasms/prevention & control , Neurodegenerative Diseases/prevention & control , Polyphenols/therapeutic use , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Gene Expression Regulation , Humans , Neoplasms/genetics , Neoplasms/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology
16.
Aging (Albany NY) ; 12(18): 18033-18051, 2020 Jul 08.
Article in English | MEDLINE | ID: mdl-32640420

ABSTRACT

Our previous studies showed that loss-of-function mutation of growth hormone releasing hormone (GHRH) results in increased longevity and enhanced insulin sensitivity in mice. However, the details of improved insulin action and tissue-specific insulin signaling are largely unknown in this healthy-aging mouse model. We conducted hyperinsulinemic-euglycemic clamp to investigate mechanisms underlying enhanced insulin sensitivity in growth hormone (GH) deficient mice. Further, we assessed in vivo tissue-specific insulin activity via activation of PI3K-AKT and MAPK-ERK1/2 cascades using western blot. Clamp results showed that the glucose infusion rate required for maintaining euglycemia was much higher in GHRH-/- mice compared to WT controls. Insulin-mediated glucose production was largely suppressed, whereas glucose uptake in skeletal muscle and brown adipose tissue were significant enhanced in GHRH-/- mice compared to WT controls. Enhanced capacity of insulin-induced activation of the PI3K-AKT and MAPK-ERK1/2 signaling were observed in a tissue-specific manner in GHRH-/- mice. Enhanced systemic insulin sensitivity in long-lived GHRH-/- mice is associated with differential activation of insulin signaling cascades among various organs. Improved action of insulin in the insulin sensitive tissues is likely to mediate the prolonged longevity and healthy-aging effects of GH deficiency in mice.

17.
Aging (Albany NY) ; 12(10): 9761-9780, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32422607

ABSTRACT

Our previous study demonstrated that the loss of growth hormone releasing hormone (GHRH) results in increased lifespan and improved metabolic homeostasis in the mouse model generated by classical embryonic stem cell-based gene-targeting method. In this study, we targeted the GHRH gene using the CRISPR/Cas9 technology to avoid passenger alleles/mutations and performed in-depth physiological and metabolic characterization. In agreement with our previous observations, male and female GHRH-/- mice have significantly reduced body weight and enhanced insulin sensitivity when compared to wild type littermates. Dual-energy X-ray absorptiometry showed that there were significant decreases in lean mass, bone mineral content and density, and a dramatic increase in fat mass of GHRH-/- mice when compared to wild type littermates. Indirect calorimetry measurements showed dramatic reductions in oxygen consumption, carbon dioxide production and energy expenditure in GHRH-/- mice compared to wild type mice in both light and dark cycles. Respiratory exchange ratio was significantly lower in GHRH-/- mice during the light cycle, but not during the dark cycle, indicating a circadian related metabolic shift towards fat utilization in the growth hormone deficient mice. The novel CRISPR/Cas9 GHRH-/- mice are exhibiting the consistent and unique physiological and metabolic characteristics, which might mediate the longevity effects of growth hormone deficiency in mice.


Subject(s)
CRISPR-Associated Protein 9/genetics , Growth Hormone-Releasing Hormone/genetics , Growth Hormone/deficiency , Longevity/genetics , Loss of Function Mutation/physiology , Absorptiometry, Photon , Animals , Body Composition/genetics , Body Weight/genetics , Calorimetry, Indirect , Energy Metabolism/genetics , Female , Insulin Resistance/genetics , Male , Mice
18.
Aging (Albany NY) ; 12(4): 3473-3485, 2020 02 23.
Article in English | MEDLINE | ID: mdl-32091406

ABSTRACT

Numerous genetic manipulations that extend lifespan in mice have been discovered over the past two decades, the most robust of which has arguably been the down regulation of growth hormone (GH) signaling. However, while decreased GH signaling has been associated with improved health and lifespan, many of the underlying physiological changes and molecular mechanisms associated with GH signaling have yet to be elucidated. To this end, we have completed the first transcriptomic and metabolomic study on long-lived growth hormone releasing hormone knockout (GHRH-KO) and wild-type mice in brown adipose tissue (transcriptomics) and blood serum (metabolomics). We find that GHRH-KO mice have increased transcript levels of mitochondrial and amino acid genes with decreased levels of extracellular matrix genes. Concurrently, mitochondrial metabolites are differentially regulated in GHRH-KO. Furthermore, we find a strong signal of genotype-by-sex interactions, suggesting the sexes have differing physiological responses to GH deficiency. Overall, our results point towards a strong influence of mitochondrial metabolism in GHRH-KO mice which potentially is tightly intertwined with their extended lifespan phenotype.


Subject(s)
Amino Acids/metabolism , Growth Hormone-Releasing Hormone/genetics , Longevity/genetics , Mitochondria/genetics , Adipose Tissue, Brown/metabolism , Animals , Growth Hormone-Releasing Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Metabolome , Metabolomics , Mice , Mice, Knockout , Mitochondria/metabolism , Phenotype , Transcriptome
19.
Proc Natl Acad Sci U S A ; 115(7): E1495-E1503, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29378959

ABSTRACT

It is well documented that inhibition of mTORC1 (defined by Raptor), a complex of mechanistic target of rapamycin (mTOR), extends life span, but less is known about the mechanisms by which mTORC2 (defined by Rictor) impacts longevity. Here, rapamycin (an inhibitor of mTOR) was used in GHR-KO (growth hormone receptor knockout) mice, which have suppressed mTORC1 and up-regulated mTORC2 signaling, to determine the effect of concurrently decreased mTORC1 and mTORC2 signaling on life span. We found that rapamycin extended life span in control normal (N) mice, whereas it had the opposite effect in GHR-KO mice. In the rapamycin-treated GHR-KO mice, mTORC2 signaling was reduced without further inhibition of mTORC1 in the liver, muscle, and s.c. fat. Glucose and lipid homeostasis were impaired, and old GHR-KO mice treated with rapamycin lost functional immune cells and had increased inflammation. In GHR-KO MEF cells, knockdown of Rictor, but not Raptor, decreased mTORC2 signaling. We conclude that drastic reduction of mTORC2 plays important roles in impaired longevity in GHR-KO mice via disruption of whole-body homeostasis.


Subject(s)
Immunosuppressive Agents/pharmacology , Longevity/drug effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Receptors, Somatotropin/physiology , Sirolimus/pharmacology , Animals , Cytoplasm/drug effects , Cytoplasm/metabolism , Female , Insulin Resistance , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction
20.
Elife ; 62017 07 04.
Article in English | MEDLINE | ID: mdl-28675141

ABSTRACT

Life-long lack of growth hormone (GH) action can produce remarkable extension of longevity in mice. Here we report that GH treatment limited to a few weeks during development influences the lifespan of long-lived Ames dwarf and normal littermate control mice in a genotype and sex-specific manner. Studies in a separate cohort of Ames dwarf mice show that this short period of the GH exposure during early development produces persistent phenotypic, metabolic and molecular changes that are evident in late adult life. These effects may represent mechanisms responsible for reduced longevity of dwarf mice exposed to GH treatment early in life. Our data suggest that developmental programming of aging importantly contributes to (and perhaps explains) the well documented developmental origins of adult disease.


Subject(s)
Growth Hormone/metabolism , Longevity , Animals , Dwarfism , Genotype , Mice , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...