1
|
Goel K, Chhetri A, Ludhiadch A, Munshi A. Current Update on Categorization of Migraine Subtypes on the Basis of Genetic Variation: a Systematic Review. Mol Neurobiol 2024; 61:4804-4833. [PMID: 38135854 DOI: 10.1007/s12035-023-03837-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
Migraine is a complex neurovascular disorder that is characterized by severe behavioral, sensory, visual, and/or auditory symptoms. It has been labeled as one of the ten most disabling medical illnesses in the world by the World Health Organization (Aagaard et al Sci Transl Med 6(237):237ra65, 2014). According to a recent report by the American Migraine Foundation (Shoulson et al Ann Neurol 25(3):252-9, 1989), around 148 million people in the world currently suffer from migraine. On the basis of presence of aura, migraine is classified into two major subtypes: migraine with aura (Aagaard et al Sci Transl Med 6(237):237ra65, 2014) and migraine without aura. (Aagaard K et al Sci Transl Med 6(237):237ra65, 2014) Many complex genetic mechanisms have been proposed in the pathophysiology of migraine but specific pathways associated with the different subtypes of migraine have not yet been explored. Various approaches including candidate gene association studies (CGAS) and genome-wide association studies (Fan et al Headache: J Head Face Pain 54(4):709-715, 2014). have identified the genetic markers associated with migraine and its subtypes. Several single nucleotide polymorphisms (Kaur et al Egyp J Neurol, Psychiatry Neurosurg 55(1):1-7, 2019) within genes involved in ion homeostasis, solute transport, synaptic transmission, cortical excitability, and vascular function have been associated with the disorder. Currently, the diagnosis of migraine is majorly behavioral with no focus on the genetic markers and thereby the therapeutic intervention specific to subtypes. Therefore, there is a need to explore genetic variants significantly associated with MA and MO as susceptibility markers in the diagnosis and targets for therapeutic interventions in the specific subtypes of migraine. Although the proper characterization of pathways based on different subtypes is yet to be studied, this review aims to make a first attempt to compile the information available on various genetic variants and the molecular mechanisms involved with the development of MA and MO. An attempt has also been made to suggest novel candidate genes based on their function to be explored by future research.
Collapse
Affiliation(s)
- Kashish Goel
- Complex Disease Genomics and Precision Medicine Laboratory, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Aakash Chhetri
- Complex Disease Genomics and Precision Medicine Laboratory, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Abhilash Ludhiadch
- Complex Disease Genomics and Precision Medicine Laboratory, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Anjana Munshi
- Complex Disease Genomics and Precision Medicine Laboratory, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India, 151401.
| |
Collapse
|
2
|
Reda GK, Ndunguru SF, Csernus B, Lugata JK, Knop R, Szabó C, Czeglédi L, Lendvai ÁZ. Sex-specific effects of dietary restriction on physiological variables in Japanese quails. Ecol Evol 2024; 14:e11405. [PMID: 38799393 PMCID: PMC11116846 DOI: 10.1002/ece3.11405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Nutritional limitation is a common phenomenon in nature that leads to trade-offs among processes competing for limited resources. These trade-offs are mediated by changes in physiological traits such as growth factors and circulating lipids. However, studies addressing the sex-specific effect of nutritional deficiency on these physiological variables are limited in birds. We used dietary restriction to mimic the depletion of resources to various degrees and investigated sex-specific effects on circulating levels of insulin-like growth factor 1 (IGF-1) and triglycerides in Japanese quails (Coturnix japonica) subjected to ad libitum, 20%, 30% or 40% restriction of their daily requirement, for 2 weeks. We also explored the association of both physiological variables with body mass and egg production. While dietary restriction showed no effects on circulating IGF-1, this hormone exhibited a marked sexual difference, with females having 64.7% higher IGF-1 levels than males. Dietary restriction significantly reduced plasma triglyceride levels in both sexes. Females showed more than six-fold higher triglyceride levels than males. Triglyceride levels were positively associated with body mass in females while showed not association in males. Overall, our findings revealed sex-specific expression of physiological variables under dietary restriction conditions, which coincide with body size.
Collapse
Affiliation(s)
- Gebrehaweria K. Reda
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental ManagementUniversity of DebrecenDebrecenHungary
- Doctoral School of Animal ScienceUniversity of DebrecenDebrecenHungary
- Department of Evolutionary Zoology and Human Biology, Faculty of Life ScienceUniversity of DebrecenDebrecenHungary
| | - Sawadi F. Ndunguru
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental ManagementUniversity of DebrecenDebrecenHungary
- Doctoral School of Animal ScienceUniversity of DebrecenDebrecenHungary
- Department of Evolutionary Zoology and Human Biology, Faculty of Life ScienceUniversity of DebrecenDebrecenHungary
| | - Brigitta Csernus
- Department of Evolutionary Zoology and Human Biology, Faculty of Life ScienceUniversity of DebrecenDebrecenHungary
| | - James K. Lugata
- Doctoral School of Animal ScienceUniversity of DebrecenDebrecenHungary
- Department of Animal Nutrition and Physiology, Faculty of Agriculture and Food Sciences and Environmental ManagementUniversity of DebrecenDebrecenHungary
| | - Renáta Knop
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental ManagementUniversity of DebrecenDebrecenHungary
| | - Csaba Szabó
- Department of Animal Nutrition and Physiology, Faculty of Agriculture and Food Sciences and Environmental ManagementUniversity of DebrecenDebrecenHungary
| | - Levente Czeglédi
- Department of Animal Science, Institute of Animal Science, Biotechnology and Nature Conservation, Faculty of Agricultural and Food Sciences and Environmental ManagementUniversity of DebrecenDebrecenHungary
| | - Ádám Z. Lendvai
- Department of Evolutionary Zoology and Human Biology, Faculty of Life ScienceUniversity of DebrecenDebrecenHungary
| |
Collapse
|
3
|
Das JK, Banskota N, Candia J, Griswold ME, Orenduff M, de Cabo R, Corcoran DL, Das SK, De S, Huffman KM, Kraus VB, Kraus WE, Martin C, Racette SB, Redman LM, Schilling B, Belsky D, Ferrucci L. Calorie restriction modulates the transcription of genes related to stress response and longevity in human muscle: The CALERIE study. Aging Cell 2023; 22:e13963. [PMID: 37823711 PMCID: PMC10726900 DOI: 10.1111/acel.13963] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 10/13/2023] Open
Abstract
The lifespan extension induced by 40% caloric restriction (CR) in rodents is accompanied by postponement of disease, preservation of function, and increased stress resistance. Whether CR elicits the same physiological and molecular responses in humans remains mostly unexplored. In the CALERIE study, 12% CR for 2 years in healthy humans induced minor losses of muscle mass (leg lean mass) without changes of muscle strength, but mechanisms for muscle quality preservation remained unclear. We performed high-depth RNA-Seq (387-618 million paired reads) on human vastus lateralis muscle biopsies collected from the CALERIE participants at baseline, 12- and 24-month follow-up from the 90 CALERIE participants randomized to CR and "ad libitum" control. Using linear mixed effect model, we identified protein-coding genes and splicing variants whose expression was significantly changed in the CR group compared to controls, including genes related to proteostasis, circadian rhythm regulation, DNA repair, mitochondrial biogenesis, mRNA processing/splicing, FOXO3 metabolism, apoptosis, and inflammation. Changes in some of these biological pathways mediated part of the positive effect of CR on muscle quality. Differentially expressed splicing variants were associated with change in pathways shown to be affected by CR in model organisms. Two years of sustained CR in humans positively affected skeletal muscle quality, and impacted gene expression and splicing profiles of biological pathways affected by CR in model organisms, suggesting that attainable levels of CR in a lifestyle intervention can benefit muscle health in humans.
Collapse
Affiliation(s)
- Jayanta Kumar Das
- Longitudinal Studies Section, Translation Gerontology BranchNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Nirad Banskota
- Computational Biology and Genomics CoreNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Julián Candia
- Longitudinal Studies Section, Translation Gerontology BranchNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | | | - Melissa Orenduff
- Duke Molecular Physiology Institute and Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Rafael de Cabo
- Translation Gerontology Branch, National Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - David L. Corcoran
- Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Sai Krupa Das
- Energy Metabolism, Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
| | - Supriyo De
- Computational Biology and Genomics CoreNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Kim Marie Huffman
- Duke Molecular Physiology Institute and Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Virginia B. Kraus
- Duke Molecular Physiology Institute and Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | - William E. Kraus
- Duke Molecular Physiology Institute and Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Corby K. Martin
- Pennington Biomedical Research CenterLouisiana State UniversityBaton RougeLouisianaUSA
| | - Susan B. Racette
- College of Health SolutionsArizona State UniversityPhoenixArizonaUSA
| | - Leanne M. Redman
- Pennington Biomedical Research CenterLouisiana State UniversityBaton RougeLouisianaUSA
| | | | - Daniel W. Belsky
- Department of Epidemiology & Butler Columbia Aging CenterColumbia University Mailman School of Public HealthNew York CityNew YorkUSA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translation Gerontology BranchNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| |
Collapse
|
4
|
Cui J, Yang Z, Wang J, Yin S, Xiao Y, Bai Y, Wang J. A cross-sectional analysis of association between visceral adiposity index and serum anti-aging protein Klotho in adults. Front Endocrinol (Lausanne) 2023; 14:1082504. [PMID: 36814582 PMCID: PMC9939517 DOI: 10.3389/fendo.2023.1082504] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/19/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND The visceral adiposity index (VAI) is regarded as a reliable indicator to assess body fat distribution and dysfunction. Klotho protein is a hormone with anti-aging biological functions. However, the relationship between them has not been researched. OBJECTS This study aimed to evaluate the association between VAI and serum anti-aging protein klotho in American adults. METHODS A cross-sectional study of participants was conducted based on the National Health and Nutrition Examination Surveys (NHANES) 2007-2016. Visceral adiposity was determined using the VAI score, while the klotho protein concentration was measured by ELISA kit. After adjusting some possible confounding variables, multivariate regression model was conducted to estimate the relationship between VAI and klotho protein. Furthermore, the smooth curve fitting and the segmented regression model were applied to examine the threshold effect and to calculate the inflection point. RESULT In total, 6 252 adults were eligible, with a mean VAI of 2.04 ± 0.03 and a mean klotho protein concentration of 848.79 ± 6.98 pg/ml. Multivariate regression analysis indicated that serum klotho protein concentration was lower in participants with high VAI score. When VAI was divided into quartiles, participants in the fourth quartiles of higher VAI had lower klotho protein levels (Q4: -32.25 pg/ml) than participants in the lowest quartile (Q1) after full adjustment (P < 0.05). Segmented regression suggested that the turning point value of VAI was 3.21. A 1-unit increase in VAI was significantly associated with lower klotho protein levels by -18.61 pg/ml (95% CI: -28.87, -8.35; P < 0.05) when VAI ranged from 0.29 to 3.21(accounting for 83.7% of the participants), however, the association was not significant when VAI ranged from 3.21 to 11.81 (P = 0.77). CONCLUSION There was a nonlinear correlation between VAI score and the serum anti-aging protein klotho concentrations, showing a saturation effect. When VAI was less than 3.21, they were negatively correlated, and when VAI was greater than 3.21, they had no obvious correlation.
Collapse
Affiliation(s)
- Jianwei Cui
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenzhen Yang
- Department of Clinical Laboratory, Nanchong Central Hospital, Nanchong, China
| | - Jiahao Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Shan Yin
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yunfei Xiao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yunjin Bai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Yunjin Bai, n; Jia Wang,
| | - Jia Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Yunjin Bai, n; Jia Wang,
| |
Collapse
|
5
|
Glucose-Related Traits and Risk of Migraine—A Potential Mechanism and Treatment Consideration. Genes (Basel) 2022; 13:genes13050730. [PMID: 35627115 PMCID: PMC9141901 DOI: 10.3390/genes13050730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/19/2022] [Accepted: 04/19/2022] [Indexed: 12/16/2022] Open
Abstract
Migraine and glucose-related (glycaemic) traits (fasting glucose, fasting insulin, and type 2 diabetes) are common and complex comorbid disorders that cause major economic and social burdens on patients and their families. Studies on the relationship between migraine and glucose-related traits have yielded inconsistent results. The purpose of this review is to synthesise and discuss the information from the available literature on the relationship between fasting glucose, fasting insulin, and type 2 diabetes (T2D) with migraine. Publications on migraine and fasting glucose, migraine and fasting insulin, and migraine and T2D were identified from a PubMed and Google Scholar database search and reviewed for this article. Multiple publications have suggested that the comorbidity of migraine and glucose-related traits may have a similar complex pathogenic mechanism, including impaired glucose homeostasis, insulin resistance, reduced cerebrovascular reactivity, abnormal brain metabolism, shared genetic factors, neurotransmitters, and sex hormones. Furthermore, several studies have found a bi-directional link between migraine with insulin resistance and T2D. There is strong evidence for a biological association between migraine headache and glucose-related traits, and burgeoning evidence for shared genetic influences. Therefore, genetic research into these comorbid traits has the potential to identify new biomarkers and therapeutic targets and provide biological insight into their relationships. We encourage healthcare professionals to consider the co-occurrence of migraine with glucose-related traits in the evaluation and treatment of their patients.
Collapse
|
6
|
Abstract
Growth hormone (GH) actions impact growth, metabolism, and body composition and have been associated with aging and longevity. Lack of GH results in slower growth, delayed maturation, and reduced body size and can lead to delayed aging, increased healthspan, and a remarkable extension of longevity. Adult body size, which is a GH-dependent trait, has a negative association with longevity in several mammalian species. Mechanistic links between GH and aging include evolutionarily conserved insulin/insulin-like growth factors and mechanistic target of rapamycin signaling pathways in accordance with long-suspected trade-offs between anabolic/growth processes and longevity. Height and the rate and regulation of GH secretion have been related to human aging, but longevity is not extended in humans with syndromes of GH deficiency or resistance. However, the risk of age-related chronic disease is reduced in individuals affected by these syndromes and various indices of increased healthspan have been reported.
Collapse
Affiliation(s)
- Andrzej Bartke
- Southern Illinois University School of Medicine, 801 N. Rutledge, P.O. Box 19628, Springfield, IL, 62794-9628, USA.
| |
Collapse
|
7
|
Zhang MY, Hu P, Feng D, Zhu YZ, Shi Q, Wang J, Zhu WY. The role of liver metabolism in compensatory-growth piglets induced by protein restriction and subsequent protein realimentation. Domest Anim Endocrinol 2021; 74:106512. [PMID: 32653740 DOI: 10.1016/j.domaniend.2020.106512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 01/31/2020] [Accepted: 06/13/2020] [Indexed: 11/21/2022]
Abstract
The aim of this work was to study the role of hepatic metabolism of compensatory growth in piglets induced by protein restriction and subsequent protein realimentation. Thirty-six weaned piglets were randomly distributed in a control group and a treatment group. The control group piglets were fed with a normal protein level diet (18.83% CP) for the entire experimental period (day 1-28). The treatment group piglets were fed with a protein-restriction diet (13.05% CP) for day 1 to day 14, and the diet was restored to normal protein level diet for day 15 to day 28. RNA-seq is used to analyze samples of liver metabolism on day 14 and day 28, respectively. Hepatic RNA-sequencing analysis revealed that some KEGG signaling pathways involved in glycolipid metabolism (eg, "AMPK signaling pathway," "insulin signaling pathway," and "glycolysis or gluconeogenesis") were significantly enriched on day 14 and day 28. On day 14, protein restriction promoted hepatic lipogenesis by increasing the genes expression level of ACACA, FASN, GAPM, and SREBP1C, decreasing protein phosphorylation levels of AMPKɑ and ACC in AMPK signaling pathway. In contrast, on day 28, protein realimentation promoted hepatic gluconeogenesis by increasing the concentration of G6Pase and PEPCK, decreasing protein phosphorylation levels of IRS1, Akt, and FoXO1 in insulin signaling pathway. In addition, protein realimentation activated the GH-IGF1 axis between the liver and skeletal muscle. Overall, these findings revealed the importance of liver metabolism in achieving compensatory growth.
Collapse
Affiliation(s)
- M Y Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - P Hu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - D Feng
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Y Z Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Q Shi
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - J Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China; National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - W Y Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China; National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
8
|
Piotrowska K, Zgutka K, Kupnicka P, Chlubek D, Pawlik A, Baranowska-Bosiacka I. Analysis of Bone Mineral Profile After Prolonged Every-Other-Day Feeding in C57BL/6J Male and Female Mice. Biol Trace Elem Res 2020; 194:177-183. [PMID: 31175634 PMCID: PMC6987084 DOI: 10.1007/s12011-019-01758-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/21/2019] [Indexed: 12/25/2022]
Abstract
Intermitted fasting or every-other-day feeding (EOD) has many positive effects in rodents and humans. Our goal was to describe how EOD influences bone mineral composition in female and male mice under prolonged EOD feeding. Male and female adult mice were fed EOD for 9 months. After this time, we used a direct method of measurement of mineral components in ashes of long bones (humerus and radius) to estimate the content of calcium (Ca), phosphorus (P), potassium (K), magnesium (Mg), and sodium (Na). We also performed histological analysis of sections of long bones. We found no significant changes in mineral composition between ad libitum and EOD fed males and females. We noted higher Ca and P contents in control males vs. females and lower content of Mg in control males vs. females. We observed the presence of marrow adipose tissue (MAT) in sections of EOD-fed females. EOD without supplementation during feeding days did not increase loss of mineral content of bones in C57BL/6J mice, but the presence of MAT only in EOD females indicates a gender-dependent response to EOD treatment in C57BL/6J mice.
Collapse
Affiliation(s)
- Katarzyna Piotrowska
- Department of Physiology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland.
| | - Katarzyna Zgutka
- Department of Physiology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Patrycja Kupnicka
- Department of Biochemistry, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry, Pomeranian Medical University in Szczecin, al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| |
Collapse
|
9
|
Scheja L, Heeren J. The endocrine function of adipose tissues in health and cardiometabolic disease. Nat Rev Endocrinol 2019; 15:507-524. [PMID: 31296970 DOI: 10.1038/s41574-019-0230-6] [Citation(s) in RCA: 397] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2019] [Indexed: 12/16/2022]
Abstract
In addition to their role in glucose and lipid metabolism, adipocytes respond differentially to physiological cues or metabolic stress by releasing endocrine factors that regulate diverse processes, such as energy expenditure, appetite control, glucose homeostasis, insulin sensitivity, inflammation and tissue repair. Both energy-storing white adipocytes and thermogenic brown and beige adipocytes secrete hormones, which can be peptides (adipokines), lipids (lipokines) and exosomal microRNAs. Some of these factors have defined targets; for example, adiponectin and leptin signal through their respective receptors that are expressed in multiple organs. For other adipocyte hormones, receptors are more promiscuous or remain to be identified. Furthermore, many of these hormones are also produced by other organs and tissues, which makes defining the endocrine contribution of adipose tissues a challenge. In this Review, we discuss the functional role of adipose tissue-derived endocrine hormones for metabolic adaptations to the environment and we highlight how these factors contribute to the development of cardiometabolic diseases. We also cover how this knowledge can be translated into human therapies. In addition, we discuss recent findings that emphasize the endocrine role of white versus thermogenic adipocytes in conditions of health and disease.
Collapse
Affiliation(s)
- Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
10
|
Hjortebjerg R, Berryman DE, Comisford R, List EO, Oxvig C, Bjerre M, Frystyk J, Kopchick JJ. Depot-specific and GH-dependent regulation of IGF binding protein-4, pregnancy-associated plasma protein-A, and stanniocalcin-2 in murine adipose tissue. Growth Horm IGF Res 2018; 39:54-61. [PMID: 29398370 DOI: 10.1016/j.ghir.2018.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 12/21/2017] [Accepted: 01/15/2018] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Pregnancy-associated plasma protein-A (PAPP-A) stimulates insulin-like growth factor (IGF)-I action through proteolytic cleavage of IGF binding protein-4 (IGFBP-4). Recently, stanniocalcin-2 (STC2) was discovered as an inhibitor of PAPP-A. Most members of the IGF system are expressed in adipose tissue (AT), but there is a relative paucity of information on the distribution of IGFBP-4, PAPP-A, and STC2 in different AT depots. Since IGF-I expression in AT is highly GH-dependent, we used bovine GH transgenic (bGH) and GH receptor knockout (GHR-/-) mice to investigate AT depot-specific expression patterns of IGFBP-4, PAPP-A, and STC2, and whether the regulation is GH-dependent. METHODS Seven-month-old male bGH, GHR-/- and wild type (WT) control mice were used. Body composition was determined, and subcutaneous, epididymal, retroperitoneal, mesenteric and brown adipose tissue (BAT) depots were collected. RNA expression of Igfbp4, Pappa, and Stc2 was assessed by reverse transcription quantitative PCR and IGFBP-4 protein by Western blotting. RESULTS Igfbp4, Pappa, and Stc2 RNA levels were differentially expressed in an AT depot-dependent manner in WT mice. Igfbp4 RNA levels were significantly higher in all white AT depots than in BAT. Pappa was most highly expressed in the mesenteric depot: levels were 7.5-fold higher in mesenteric than in subcutaneous AT (p < .001). Although intraabdominal in origin, epididymal and retroperitoneal Pappa expression levels were 69% and 68% lower, respectively, as compared to mesenteric levels (p < .001). Stc2 RNA expression was significantly higher in all intraabdominal white AT as compared to subcutaneous AT and BAT; levels in epididymal, retroperitoneal, and mesenteric were all more than three-fold higher than in subcutaneous AT (p < .001) and 12-fold higher than in BAT (p < .001). Gene expression patterns in bGH and GHR-/- mice mimicked those in WT mice, suggesting that GH does not affect the transcription of the STC2-PAPP-A-IGFBP-4-axis in AT. However, proteins levels of intact IGFBP-4 were significantly increased in bGH mice and decreased in GHR-/- mice, whereas the PAPP-A-generated IGFBP-4 fragment level was unaltered. CONCLUSION Expression of Igfbp4, Pappa, and Stc2 differ between AT depots and is generally higher in white AT than in BAT. The transcription appears to occur in a GH-independent manner, whereas IGFBP-4 protein levels are highly influenced by altered GH activity.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark; Danish Diabetes Academy, Odense, Denmark; Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; The Diabetes Institute, Ohio University, Athens, OH 45701, USA.
| | - Ross Comisford
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; The Diabetes Institute, Ohio University, Athens, OH 45701, USA.
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; The Diabetes Institute, Ohio University, Athens, OH 45701, USA.
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Faculty of Science and Technology, Aarhus University, Aarhus, Denmark.
| | - Mette Bjerre
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark.
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark; Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark.
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; The Diabetes Institute, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
11
|
Victoria B, Nunez Lopez YO, Masternak MM. MicroRNAs and the metabolic hallmarks of aging. Mol Cell Endocrinol 2017; 455:131-147. [PMID: 28062199 PMCID: PMC5724961 DOI: 10.1016/j.mce.2016.12.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 10/10/2016] [Accepted: 12/16/2016] [Indexed: 12/12/2022]
Abstract
Aging, the natural process of growing older, is characterized by a progressive deterioration of physiological homeostasis at the cellular, tissue, and organismal level. Metabolically, the aging process is characterized by extensive changes in body composition, multi-tissue/multi-organ insulin resistance, and physiological declines in multiple signaling pathways including growth hormone, insulin/insulin-like growth factor 1, and sex steroids regulation. With this review, we intend to consolidate published information about microRNAs that regulate critical metabolic processes relevant to aging. In certain occasions we uncover relationships likely relevant to aging, which has not been directly described before, such as the miR-451/AMPK axis. We have also included a provocative section highlighting the potential role in aging of a new designation of miRNAs, namely fecal miRNAs, recently discovered to regulate intestinal microbiota in mammals.
Collapse
Affiliation(s)
- Berta Victoria
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, USA.
| | - Yury O Nunez Lopez
- Translational Research Institute for Metabolism & Diabetes. Florida Hospital, 301 East Princeton St, Orlando, FL 32804, USA.
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, USA; Department of Head and Neck Surgery, The Greater Poland Cancer Centre, 15 Garbary St., 61-866, Poznan, Poland.
| |
Collapse
|
12
|
Gesing A, Wiesenborn D, Do A, Menon V, Schneider A, Victoria B, Stout MB, Kopchick JJ, Bartke A, Masternak MM. A Long-lived Mouse Lacking Both Growth Hormone and Growth Hormone Receptor: A New Animal Model for Aging Studies. J Gerontol A Biol Sci Med Sci 2017; 72:1054-1061. [PMID: 27688483 PMCID: PMC5861925 DOI: 10.1093/gerona/glw193] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 09/13/2016] [Indexed: 11/14/2022] Open
Abstract
Disruption of the growth hormone (GH) signaling pathway promotes insulin sensitivity and is associated with both delayed aging and extended longevity. Two kinds of long-lived mice-Ames dwarfs (df/df) and GH receptor gene-disrupted knockouts (GHRKO) are characterized by a suppressed GH axis with a significant reduction of body size and decreased plasma insulin-like growth factor-1 (IGF-1) and insulin levels. Ames dwarf mice are deficient in GH, prolactin, and thyrotropin, whereas GHRKOs are GH resistant and are dwarf with decreased circulating IGF-1 and increased GH. Crossing Ames dwarfs and GHRKOs produced a new mouse line (df/KO), lacking both GH and GH receptor. These mice are characterized by improved glucose tolerance and increased adiponectin level, which could imply that these mice should be also characterized by additional life-span extension when comparing with GHRKOs and Ames dwarfs. Importantly, our longevity experiments showed that df/KO mice maintain extended longevity when comparing with N control mice; however, they do not live longer than GHRKO and Ames df/df mice. These important findings indicate that silencing GH signal is important to extend the life span; however, further decrease of body size in mice with already inhibited GH signal does not extend the life span regardless of improved some health-span markers.
Collapse
Affiliation(s)
- Adam Gesing
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield
- Department of Oncological Endocrinology, Medical University of Lodz, Poland
| | - Denise Wiesenborn
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
- Department of Medical Biochemistry and Molecular Biology, University of Saarland, Homburg, Germany
- Department of Biotechnology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Andrew Do
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton
| | - Vinal Menon
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia
| | - Augusto Schneider
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rio Grande do Sul, Brazil
| | - Berta Victoria
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
| | - Michael B Stout
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens
| | - Andrzej Bartke
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando
- Department of Head and Neck Surgery, The Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|
13
|
Hjortebjerg R, Berryman DE, Comisford R, Frank SJ, List EO, Bjerre M, Frystyk J, Kopchick JJ. Insulin, IGF-1, and GH Receptors Are Altered in an Adipose Tissue Depot-Specific Manner in Male Mice With Modified GH Action. Endocrinology 2017; 158:1406-1418. [PMID: 28323915 PMCID: PMC5460824 DOI: 10.1210/en.2017-00084] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 02/22/2017] [Indexed: 12/28/2022]
Abstract
Growth hormone (GH) is a determinant of glucose homeostasis and adipose tissue (AT) function. Using 7-month-old transgenic mice expressing the bovine growth hormone (bGH) gene and growth hormone receptor knockout (GHR-/-) mice, we examined whether changes in GH action affect glucose, insulin, and pyruvate tolerance and AT expression of proteins involved in the interrelated signaling pathways of GH, insulinlike growth factor 1 (IGF-1), and insulin. Furthermore, we searched for AT depot-specific differences in control mice. Glycated hemoglobin levels were reduced in bGH and GHR-/- mice, and bGH mice displayed impaired gluconeogenesis as judged by pyruvate tolerance testing. Serum IGF-1 was elevated by 90% in bGH mice, whereas IGF-1 and insulin were reduced by 97% and 61% in GHR-/- mice, respectively. Igf1 RNA was increased in subcutaneous, epididymal, retroperitoneal, and brown adipose tissue (BAT) depots in bGH mice (mean increase ± standard error of the mean in all five depots, 153% ± 27%) and decreased in all depots in GHR-/- mice (mean decrease, 62% ± 4%). IGF-1 receptor expression was decreased in all AT depots of bGH mice (mean decrease, 49% ± 6%) and increased in all AT depots of GHR-/- mice (mean increase, 94% ± 8%). Insulin receptor expression was reduced in retroperitoneal, mesenteric, and BAT depots in bGH mice (mean decrease in all depots, 56% ± 4%) and augmented in subcutaneous, retroperitoneal, mesenteric, and BAT depots in GHR-/- mice (mean increase: 51% ± 1%). Collectively, our findings indicate a role for GH in influencing hormone signaling in AT in a depot-dependent manner.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
- Danish Diabetes Academy, 5000 Odense, Denmark
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
| | - Darlene E. Berryman
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701
- The Diabetes Institute at Ohio University, Ohio University, Athens, Ohio 45701
| | - Ross Comisford
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- The Diabetes Institute at Ohio University, Ohio University, Athens, Ohio 45701
| | - Stuart J. Frank
- Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35924
- Medical Service, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama 35233
| | - Edward O. List
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
| | - Mette Bjerre
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, 8000 Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, 8000 Aarhus, Denmark
| | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701
| |
Collapse
|
14
|
Martyniak K, Masternak MM. Changes in adipose tissue cellular composition during obesity and aging as a cause of metabolic dysregulation. Exp Gerontol 2016; 94:59-63. [PMID: 27939445 DOI: 10.1016/j.exger.2016.12.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/02/2016] [Accepted: 12/07/2016] [Indexed: 12/17/2022]
Abstract
Adipose tissue represents complex endocrine organ containing several different cellular populations including adipocytes, pre-adipocytes, mesenchymal stem cells, macrophages and lymphocytes. It is well establishing that these populations are not static but alter during obesity and aging. Changes in cellular populations alter inflammatory status and other common metabolic complications arise, therefore adipose tissue cellular composition helps dictate its endocrine and regulatory function. During excessive weight gain in obese individuals and as we age there is shift towards increase populations of inflammatory macrophages with a decrease of regulatory T cell. This altered cellular composition promote chronic low grade inflammation negatively affecting mesenchymal stem cell progenitor self-renewal, which result in deterioration of adipogenesis and increased cellular stress in adipocytes. All these changes promote metabolic disorders including age- or obese-related insulin resistance leading to type 2 diabetes.
Collapse
Affiliation(s)
- Kari Martyniak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, United States
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, United States; Department of Head and Neck Surgery, The Greater Poland Cancer Centre, Poznan, Poland.
| |
Collapse
|
15
|
Rocha JS, Bonkowski MS, França LR, Bartke A. Mild Calorie Restriction Does Not Affect Testosterone Levels and Testicular Gene Expression in Mutant Mice. Exp Biol Med (Maywood) 2016; 232:1050-63. [PMID: 17720951 DOI: 10.3181/0703-rm-52] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The hypothalamic-pituitary-gonadal (HPG) axis and the somatotropic axis are influenced by nutritional factors. Calorie restriction (CR) extends lifespan but suppresses both the HPG and the somatotropic axes. Since most CR studies use a fairly severe (40%–60%) reduction of calorie intake, we hypothesized that a milder CR (20%) might not be deleterious to reproduction in male mice. To test this hypothesis, we evaluated the effects of 20% CR on testicular testosterone content and on testicular expression of genes that are relevant to testicular function and reproductive competence, including insulin-like growth factor-I, cytochrome P450 aromatase (Cyp19a1), androgen receptor, luteinizing hormone receptor, follicle-stimulating hormone receptor, cytochrome P450c17 and 3-β-hydroxysteroid dehydrogenase/isomerase. To relate CR effects to the activity of the somatotropic axis, we have used growth hormone–resistant GHR knockout mice as well as transgenic mice overexpressing GH. Mild CR did not affect testosterone levels in testis homogenates and had little effect on expression of the examined genes in the reproductive organs. Altered activity of the GH/insulin-like growth factor–1 axis had a major impact on the parameters analyzed. The results also suggest that expression of several key genes involved in the control of testicular function is preserved under conditions of mild CR and encourage speculation that mild regimens of CR can produce longevity benefits without impairing reproduction.
Collapse
Affiliation(s)
- Juliana S Rocha
- Laboratory of Cellular Biology, Avenue Antonio Carlos 6627, Pampulha Belo Horizonte, MG 31270-901, Brazil.
| | | | | | | |
Collapse
|
16
|
Victoria B, Dhahbi JM, Nunez Lopez YO, Spinel L, Atamna H, Spindler SR, Masternak MM. Circulating microRNA signature of genotype-by-age interactions in the long-lived Ames dwarf mouse. Aging Cell 2015; 14:1055-66. [PMID: 26176567 PMCID: PMC4693471 DOI: 10.1111/acel.12373] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2015] [Indexed: 11/29/2022] Open
Abstract
Recent evidence demonstrates that serum levels of specific miRNAs significantly change with age. The ability of circulating sncRNAs to act as signaling molecules and regulate a broad spectrum of cellular functions implicates them as key players in the aging process. To discover circulating sncRNAs that impact aging in the long‐lived Ames dwarf mice, we conducted deep sequencing of small RNAs extracted from serum of young and old mice. Our analysis showed genotype‐specific changes in the circulating levels of 21 miRNAs during aging [genotype‐by‐age interaction (GbA)]. Genotype‐by‐age miRNAs showed four distinct expression patterns and significant overtargeting of transcripts involved in age‐related processes. Functional enrichment analysis of putative and validated miRNA targets highlighted cellular processes such as tumor suppression, anti‐inflammatory response, and modulation of Wnt, insulin, mTOR, and MAPK signaling pathways, among others. The comparative analysis of circulating GbA miRNAs in Ames mice with circulating miRNAs modulated by calorie restriction (CR) in another long‐lived mouse suggests CR‐like and CR‐independent mechanisms contributing to longevity in the Ames mouse. In conclusion, we showed for the first time a signature of circulating miRNAs modulated by age in the long‐lived Ames mouse.
Collapse
Affiliation(s)
- Berta Victoria
- Burnett School of Biomedical Sciences College of Medicine University of Central Florida 6900 Lake Nona Blvd. Orlando FL 32827 USA
| | - Joseph M. Dhahbi
- Department of Biochemistry University of California at Riverside Riverside CA 92521 USA
- Center for Genetics Childrens Hospital Oakland Research Institute Oakland CA 94609 USA
| | - Yury O. Nunez Lopez
- Translational Research Institute for Metabolism and Diabetes Florida Hospital 301 E. Princeton Street Orlando FL 2804 USA
| | - Lina Spinel
- Burnett School of Biomedical Sciences College of Medicine University of Central Florida 6900 Lake Nona Blvd. Orlando FL 32827 USA
| | - Hani Atamna
- Department of Medical Education California Northstate University Elk Grove CA USA
| | - Stephen R. Spindler
- Department of Biochemistry University of California at Riverside Riverside CA 92521 USA
| | - Michal M. Masternak
- Burnett School of Biomedical Sciences College of Medicine University of Central Florida 6900 Lake Nona Blvd. Orlando FL 32827 USA
- Department of Head and Neck Surgery The Greater Poland Cancer Centre 15 Garbary St. 61‐866 Poznan Poland
| |
Collapse
|
17
|
Villegas R, Delahanty R, Williams S, Li H, O'Brian R, Shi J, Cai Q, Xiang YB, Shu XO. Genetic Variation and Insulin Resistance in Middle-Aged Chinese Men. Ann Hum Genet 2015; 79:357-365. [PMID: 26252243 PMCID: PMC4949159 DOI: 10.1111/ahg.12124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 04/09/2015] [Indexed: 12/27/2022]
Abstract
We investigated the effect of variants in the first three genes in the insulin signaling pathway and genes identified from genome wide association studies (GWAS) of T2D quantitative traits with IR (fasting insulin and the homeostasis model assessment of IR, HOMA-IR) and evaluated gene-environment interactions with IR traits among 1879 nondiabetic middle-aged men from a population-based study conducted in Shanghai, China. One candidate gene, IGF1, was associated with fasting insulin and HOMA-IR. We observed four BMI-gene interactions (P < 0.05) with HOMA-IR (INRS rs7254060, INRS rs7254358, GLU4 rs2113050, and GLU4 rs7713127) and seven BMI-gene interactions with fasting insulin (INRS rs7254060, INRS rs7254358, INRS rs10417205, INRS rs1799817, GLU4 rs12054720 GLU4 rs2113050, and GLU4 rs7713127). There were four WHR-gene interactions with HOMA-IR (INRS rs10417205, INRS rs12971499, INRS rs7254060, and INRS rs7254358), five WHR-gene interactions with fasting insulin (INRS rs10417205, INRS rs7254060, INRS rs7254358, GLU4 rs2113050, and GLU4 rs7713127), eight physical activity-gene interactions with HOMA-IR (INRS rs10411676, INRS rs11671297, INRS rs2229431, INRS rs12461909, INRS rs6510950, INRS rs10420382, IRS2 rs913949, and IRS2 rs2241745) and five physical activity-gene interactions with fasting insulin (INRS rs2229431, INRS rs12461909, INRS rs10420382, IRS2 rs913949, and IRS2 rs2241745). Our results suggest that BMI, WHR and physical activity may modify IR-associated variants.
Collapse
Affiliation(s)
- Raquel Villegas
- Vanderbilt Epidemiology Center, Department of Medicine, Vanderbilt University Medical Center, TN, USA
| | - Ryan Delahanty
- Vanderbilt Epidemiology Center, Department of Medicine, Vanderbilt University Medical Center, TN, USA
| | - Scott Williams
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Honglan Li
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, P.R. China
| | - Richard O'Brian
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jiajun Shi
- Vanderbilt Epidemiology Center, Department of Medicine, Vanderbilt University Medical Center, TN, USA
| | - Qiuyin Cai
- Vanderbilt Epidemiology Center, Department of Medicine, Vanderbilt University Medical Center, TN, USA
| | - Yong-Bing Xiang
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, P.R. China
| | - Xiao Ou Shu
- Vanderbilt Epidemiology Center, Department of Medicine, Vanderbilt University Medical Center, TN, USA
| |
Collapse
|
18
|
Wiesenborn DS, Menon V, Zhi X, Do A, Gesing A, Wang Z, Bartke A, Altomare DA, Masternak MM. The effect of calorie restriction on insulin signaling in skeletal muscle and adipose tissue of Ames dwarf mice. Aging (Albany NY) 2015; 6:900-12. [PMID: 25411241 PMCID: PMC4247389 DOI: 10.18632/aging.100700] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Long-living Ames dwarf (df/df) mice are homozygous for a mutation of the Prop1(df) gene. As a result, mice are deficient in growth hormone (GH), prolactin (PRL) and thyrotropin (TSH). In spite of the hormonal deficiencies, df/df mice live significantly longer and healthier lives compared to their wild type siblings. We studied the effects of calorie restriction (CR) on the expression of insulin signaling genes in skeletal muscle and adipose tissue of normal and df/df mice. The analysis of genes expression showed that CR differentially affects the insulin signaling pathway in these insulin target organs. Moreover, results obtained in both normal and Ames dwarf mice indicate more direct effects of CR on insulin signaling genes in adipose tissue than in skeletal muscle. Interestingly, CR reduced the protein levels of adiponectin in the epididymal adipose tissue of normal and Ames dwarf mice, while elevating adiponectin levels in skeletal muscle and plasma of normal mice only. In conclusion, our findings suggest that both skeletal muscle and adipose tissue are important mediators of insulin effects on longevity. Additionally, the results revealed divergent effects of CR on expression of genes in the insulin signaling pathway of normal and Ames dwarf mice.
Collapse
Affiliation(s)
- Denise S Wiesenborn
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Vinal Menon
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA. Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina Columbia, SC 29209, USA
| | - Xu Zhi
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA. Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Andrew Do
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Adam Gesing
- Department of Oncological Endocrinology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Zhihui Wang
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Deborah A Altomare
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA. Department of Head and Neck Surgery, The Greater Poland Cancer Centre, 61-866 Poznan
| |
Collapse
|
19
|
Zawada I, Masternak MM, List EO, Stout MB, Berryman DE, Lewinski A, Kopchick JJ, Bartke A, Karbownik-Lewinska M, Gesing A. Gene expression of key regulators of mitochondrial biogenesis is sex dependent in mice with growth hormone receptor deletion in liver. Aging (Albany NY) 2015; 7:195-204. [PMID: 25855408 PMCID: PMC4394730 DOI: 10.18632/aging.100733] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 03/24/2015] [Indexed: 12/29/2022]
Abstract
Mitochondrial biogenesis is an essential process for cell viability. Mice with disruption of the growth hormone receptor (GHR) gene (Ghr gene) in the liver (LiGHRKO), in contrast to long-lived mice with global deletion of the Ghr gene (GHRKO), are characterized by lack of improved insulin sensitivity and severe hepatic steatosis. Tissue-specific disruption of the GHR in liver results in a mouse model with dramatically altered GH/IGF1 axis. We have previously shown increased levels of key regulators of mitochondrial biogenesis in insulin-sensitive GHRKO mice. The aim of the present study is to assess, using real-time PCR, the gene expression of key regulators of mitochondrial biogenesis (Pgc1α, Ampk, Sirt1, Nrf2 and Mfn2) and a marker of mitochondrial activity (CoxIV) in brains, kidneys and livers of male and female LiGHRKO and wild-type (WT) mice. There were significant differences between males and females. In the brain, expression of Pgc1α, Ampk, Sirt1, Nrf2 and Mfn2 was lower in pooled females compared to pooled males. In the kidneys, expression of Ampk and Sirt1 was also lower in female mice. In the liver, no differences between males and females were observed. Sexual dimorphism may play an important role in regulating the biogenesis of mitochondria.
Collapse
Affiliation(s)
- Ilona Zawada
- Department of Oncological Endocrinology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Michal M. Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, USA
- Department of Head and Neck Surgery, The Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Edward O. List
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
- Department of Specialty Medicine, Ohio University, Athens, OH 45701, USA
| | - Michael B. Stout
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Darlene E. Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
- School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Andrzej Lewinski
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, 93-338 Lodz, Poland
- Department of Endocrinology and Metabolic Diseases, Polish Mother's Memorial Hospital – Research Institute, 93-338 Lodz, Poland
| | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Malgorzata Karbownik-Lewinska
- Department of Oncological Endocrinology, Medical University of Lodz, 90-752 Lodz, Poland
- Department of Endocrinology and Metabolic Diseases, Polish Mother's Memorial Hospital – Research Institute, 93-338 Lodz, Poland
| | - Adam Gesing
- Department of Oncological Endocrinology, Medical University of Lodz, 90-752 Lodz, Poland
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| |
Collapse
|
20
|
Growth hormone abolishes beneficial effects of calorie restriction in long-lived Ames dwarf mice. Exp Gerontol 2014; 58:219-229. [PMID: 25152388 DOI: 10.1016/j.exger.2014.08.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/16/2014] [Accepted: 08/20/2014] [Indexed: 11/23/2022]
Abstract
Disruption of the growth hormone (GH) axis promotes longevity and delays aging. In contrast, GH over-expression may lead to accelerated aging and shorter life. Calorie restriction (CR) improves insulin sensitivity and may extend lifespan. Long-lived Ames dwarf (df/df) mice have additional extension of longevity when subjected to 30% CR. The aim of the study was to assess effects of CR or GH replacement therapy separately and as a combined (CR+GH) treatment in GH-deficient df/df and normal mice, on selected metabolic parameters (e.g., insulin, glucose, cholesterol), insulin signaling components (e.g., insulin receptor [IR] β-subunit, phosphorylated form of IR [IR pY1158], protein kinase C ζ/λ [p-PKCζ/λ] and mTOR [p-mTOR]), transcription factor p-CREB, and components of the mitogen-activated protein kinase (MAPK) signaling (p-ERK1/2, p-p38), responsible for cell proliferation, differentiation and survival. CR decreased plasma levels of insulin, glucose, cholesterol and leptin, and increased hepatic IR β-subunit and IR pY1158 levels as well as IR, IRS-1 and GLUT-2 gene expression compared to ad libitum feeding, showing a significant beneficial diet intervention effect. Moreover, hepatic protein levels of p-PKCζ/λ, p-mTOR and p-p38 decreased, and p-CREB increased in CR mice. On the contrary, GH increased levels of glucose, cholesterol and leptin in plasma, and p-mTOR or p-p38 in livers, and decreased plasma adiponectin and hepatic IR β-subunit compared to saline treatment. There were no GH effects on adiponectin in N mice. Moreover, GH replacement therapy did not affect IR, IRS-1 and GLUT-2 gene expression. GH treatment abolishes the beneficial effects of CR; it may suggest an important role of GH-IGF1 axis in mediating the CR action. Suppressed somatotrophic signaling seems to predominate over GH replacement therapy in the context of the examined parameters and signaling pathways.
Collapse
|
21
|
Zabuga OG, Akhaladze NG, Vaiserman AM. Nutritional programming: Theoretical concepts and experimental evidence. ADVANCES IN GERONTOLOGY 2014. [DOI: 10.1134/s2079057014010159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Gesing A, Wang F, List EO, Berryman DE, Masternak MM, Lewinski A, Karbownik-Lewinska M, Kopchick JJ, Bartke A. Expression of apoptosis-related genes in liver-specific growth hormone receptor gene-disrupted mice is sex dependent. J Gerontol A Biol Sci Med Sci 2014; 70:44-52. [PMID: 24550353 DOI: 10.1093/gerona/glu008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Apoptosis is a process that affects life span and health. Mice with liver-specific disruption of the growth hormone receptor (GHR) gene (ie, Ghr gene) liver-specific growth hormone receptor knockout [LiGHRKO] mice), as opposed to mice with global deletion of the Ghr gene (GHRKO; Ghr-/-), are characterized by severe hepatic steatosis and lack of improved insulin sensitivity. We have previously shown that levels of proapoptotic factors are decreased in long-lived and insulin-sensitive GHRKO mice. In the current study, expression of specific apoptosis-related genes was assessed in brains, kidneys, and livers of male and female LiGHRKO and wild-type mice using real-time PCR. In the brain, expression of Caspase 3, Caspase 9, Smac/DIABLO, and p53 was decreased in females compared with males. Renal expression of Caspase 3 and Noxa also decreased in female mice. In the liver, no differences were seen between males and females. Also, no significant genotype effects were detected in the examined organs. Lack of significant genotype effect in kidneys contrasts with previous observations in GHRKO mice. Apparently, global GHR deletion induces beneficial changes in apoptotic factors, whereas liver-specific GHR disruption does not. Furthermore, sexual dimorphism may play an important role in regulating apoptosis during liver-specific suppression of the somatotrophic signaling.
Collapse
Affiliation(s)
- Adam Gesing
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield. Department of Oncological Endocrinology, Medical University of Lodz, Poland.
| | - Feiya Wang
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield
| | - Edward O List
- Edison Biotechnology Institute, Department of Specialty Medicine
| | - Darlene E Berryman
- Edison Biotechnology Institute, School of Applied Health Sciences and Wellness, and Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando. Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Andrzej Lewinski
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, Poland. Department of Endocrinology and Metabolic Diseases, Polish Mother's Memorial Hospital - Research Institute, Lodz, Poland
| | - Malgorzata Karbownik-Lewinska
- Department of Oncological Endocrinology, Medical University of Lodz, Poland. Department of Endocrinology and Metabolic Diseases, Polish Mother's Memorial Hospital - Research Institute, Lodz, Poland
| | - John J Kopchick
- Edison Biotechnology Institute, Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens
| | - Andrzej Bartke
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield
| |
Collapse
|
23
|
Sackmann-Sala L, Berryman DE, Lubbers ER, Zhang H, Vesel CB, Troike KM, Gosney ES, List EO, Kopchick JJ. Age-related and depot-specific changes in white adipose tissue of growth hormone receptor-null mice. J Gerontol A Biol Sci Med Sci 2013; 69:34-43. [PMID: 23873966 DOI: 10.1093/gerona/glt110] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
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.
Collapse
Affiliation(s)
- Lucila Sackmann-Sala
- Edison Biotechnology Institute, Ohio University, 1 Water Tower Dr., The Ridges, Athens, OH 45701.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ding J, Sackmann-Sala L, Kopchick JJ. Mouse models of growth hormone action and aging: a proteomic perspective. Proteomics 2012; 13:674-85. [PMID: 23019135 DOI: 10.1002/pmic.201200271] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 07/19/2012] [Accepted: 07/27/2012] [Indexed: 12/12/2022]
Abstract
Growth hormone (GH) is a protein secreted by the anterior pituitary and circulates throughout the body to exert important actions on growth and metabolism. GH stimulates the secretion of insulin-like growth factor-I (IGF-I) that mediates some of the growth promoting actions of GH. The GH/IGF-I axis has recently been recognized as important in terms of longevity in organisms ranging from Caenorhabditis elegans to mice. For example, GH transgenic mice possess short lifespans while GH receptor null (GHR-/-) mice have extended longevity. Thus, the actions of GH (or IGF-I) or lack thereof impact the aging process. In this review, we summarize the proteomic analyses of plasma and white adipose tissue in these two mouse models of GH action, i.e. GH transgenic and GHR-/- mice. At the protein level, we wanted to establish novel plasma biomarkers of GH action as a function of age and to determine differences in adipose tissue depots. We have shown that these proteomic approaches have not only confirmed several known physiological actions of GH, but also resulted in novel protein biomarkers and targets that may be indicative of the aging process and/or new functions of GH. These results may generate new directions for GH and/or aging research.
Collapse
Affiliation(s)
- Juan Ding
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
25
|
Ghrelin improves body weight loss and skeletal muscle catabolism associated with angiotensin II-induced cachexia in mice. ACTA ACUST UNITED AC 2012; 178:21-8. [PMID: 22750276 DOI: 10.1016/j.regpep.2012.06.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 05/25/2012] [Accepted: 06/20/2012] [Indexed: 12/20/2022]
Abstract
Ghrelin is a gastric peptide that regulates energy homeostasis. Angiotensin II (Ang II) is known to induce body weight loss and skeletal muscle catabolism through the ubiquitin-proteasome pathway. In this study, we investigated the effects of ghrelin on body weight and muscle catabolism in mice treated with Ang II. The continuous subcutaneous administration of Ang II to mice for 6 days resulted in cardiac hypertrophy and significant decreases in body weight gain, food intake, food efficiency, lean mass, and fat mass. In the gastrocnemius muscles of Ang II-treated mice, the levels of insulin-like growth factor 1 (IGF-1) were decreased, and the levels of mRNA expression of catabolic factors were increased. Although the repeated subcutaneous injections of ghrelin (1.0mg/kg, twice daily for 5 days) did not affect cardiac hypertrophy, they resulted in significant body weight gains and improved food efficiencies and tended to increase both lean and fat mass in Ang II-treated mice. Ghrelin also ameliorated the decreased IGF-1 levels and the increased mRNA expression levels of catabolic factors in the skeletal muscle. IGF-1 mRNA levels in the skeletal muscle significantly decreased 24h after Ang II infusion, and this was reversed by two subcutaneous injections of ghrelin. In C2C12-derived myocytes, the dexamethasone-induced mRNA expression of atrogin-1 was decreased by IGF-1 but not by ghrelin. In conclusion, we demonstrated that ghrelin improved body weight loss and skeletal muscle catabolism in mice treated with Ang II, possibly through the early restoration of IGF-1 mRNA in the skeletal muscle and the amelioration of nutritional status.
Collapse
|
26
|
Softic S, Kirby M, Berger NG, Shroyer NF, Woods SC, Kohli R. Insulin concentration modulates hepatic lipid accumulation in mice in part via transcriptional regulation of fatty acid transport proteins. PLoS One 2012; 7:e38952. [PMID: 22745692 PMCID: PMC3380053 DOI: 10.1371/journal.pone.0038952] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 05/16/2012] [Indexed: 12/18/2022] Open
Abstract
Background Fatty liver disease (FLD) is commonly associated with insulin resistance and obesity, but interestingly it is also observed at low insulin states, such as prolonged fasting. Thus, we asked whether insulin is an independent modulator of hepatic lipid accumulation. Methods/Principal Findings In mice we induced, hypo- and hyperinsulinemia associated FLD by diet induced obesity and streptozotocin treatment, respectively. The mechanism of free fatty acid induced steatosis was studied in cell culture with mouse liver cells under different insulin concentrations, pharmacological phosphoinositol-3-kinase (PI3K) inhibition and siRNA targeted gene knock-down. We found with in vivo and in vitro models that lipid storage is increased, as expected, in both hypo- and hyperinsulinemic states, and that it is mediated by signaling through either insulin receptor substrate (IRS) 1 or 2. As previously reported, IRS-1 was up-regulated at high insulin concentrations, while IRS-2 was increased at low levels of insulin concentration. Relative increase in either of these insulin substrates, was associated with an increase in liver-specific fatty acid transport proteins (FATP) 2&5, and increased lipid storage. Furthermore, utilizing pharmacological PI3K inhibition we found that the IRS-PI3K pathway was necessary for lipogenesis, while FATP responses were mediated via IRS signaling. Data from additional siRNA experiments showed that knock-down of IRSs impacted FATP levels. Conclusions/Significance States of perturbed insulin signaling (low-insulin or high-insulin) both lead to increased hepatic lipid storage via FATP and IRS signaling. These novel findings offer a common mechanism of FLD pathogenesis in states of both inadequate (prolonged fasting) and ineffective (obesity) insulin signaling.
Collapse
Affiliation(s)
- Samir Softic
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, Riley Hospital for Children, Indiana University, Indianapolis, Indiana, United States of America
| | - Michelle Kirby
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Nicholas G. Berger
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Noah F. Shroyer
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Stephen C. Woods
- Metabolic Diseases Institute, Obesity Research Center, Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Rohit Kohli
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
27
|
Ko F, Yu Q, Xue QL, Yao W, Brayton C, Yang H, Fedarko N, Walston J. Inflammation and mortality in a frail mouse model. AGE (DORDRECHT, NETHERLANDS) 2012; 34:705-15. [PMID: 21633802 PMCID: PMC3337927 DOI: 10.1007/s11357-011-9269-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Accepted: 05/09/2011] [Indexed: 05/02/2023]
Abstract
Mice homozygous for targeted deletion of the interleukin 10 gene (Il-10) have been partially characterized as a model for human frailty. These mice have increased serum interleukin (IL)-6 in midlife, skeletal muscle weakness, and an altered skeletal muscle gene expression profile compared to age and sex-matched C57BL/6 (B6) control mice. In order to further characterize for use as a frailty model, we evaluated the evolution of inflammatory pathway activation, endocrine change, and mortality in these mice. Serum was collected in groups of age- and sex-matched B6.129P2-Il10(tm1Cgn)/J (IL-10(tm/tm)) mice and B6 control mice at age 12, 24, 48, 72, and 90 weeks. Cytokines including IL-6, interleukin 1 beta (IL-1β), tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), chemokine (C-X-C motif) ligand 1 (KC), IL-12, and IL-10 were measured using electro-chemiluminescent multiplex immunoassay and insulin-like growth factor 1 (IGF-1) was measured using solid-phase enzyme-linked immunosorbent assay. A separate longitudinal cohort was monitored from age 35 weeks to approximately 100 weeks. Survival was evaluated by Kaplan-Meier survival estimates and detailed necropsy information was gathered in a subset of mice that died or were sacrificed. In IL-10(tm/tm) mice compared to B6 controls, serum IL-6, IL-1β, TNF-α, IFN-γ, KC levels were significantly elevated across the age groups, serum mean IGF-1 levels were higher in the 48-week-old groups, and overall mortality rate was significantly higher. The quadratic relationship between IGF-1 and age was significantly different between the two strains of mice. Serum IL-6 was positively associated with IGF-1 but the effect was significantly larger in IL-10(tm/tm) mice. These findings provide additional rationale for the use of the IL-10(tm/tm) mouse as a model for frailty and for low-grade inflammatory pathway activation.
Collapse
Affiliation(s)
- Fred Ko
- Brookdale Department of Geriatrics and Palliative Medicine, Mount Sinai School of Medicine, New York, NY 10029 USA
| | - Qilu Yu
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD 21224 USA
| | - Qian-Li Xue
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD 21224 USA
| | - Wenliang Yao
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD 21224 USA
| | - Cory Brayton
- Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Huanle Yang
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD 21224 USA
| | - Neal Fedarko
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD 21224 USA
| | - Jeremy Walston
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD 21224 USA
- Johns Hopkins Asthma and Allergy Center, Suite 1A.62, 5501 Bayview Circle, Baltimore, MD 21224 USA
| |
Collapse
|
28
|
Duarte AI, Moreira PI, Oliveira CR. Insulin in central nervous system: more than just a peripheral hormone. J Aging Res 2012; 2012:384017. [PMID: 22500228 PMCID: PMC3303591 DOI: 10.1155/2012/384017] [Citation(s) in RCA: 217] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 10/12/2011] [Accepted: 11/23/2011] [Indexed: 12/14/2022] Open
Abstract
Insulin signaling in central nervous system (CNS) has emerged as a novel field of research since decreased brain insulin levels and/or signaling were associated to impaired learning, memory, and age-related neurodegenerative diseases. Thus, besides its well-known role in longevity, insulin may constitute a promising therapy against diabetes- and age-related neurodegenerative disorders. More interestingly, insulin has been also faced as the potential missing link between diabetes and aging in CNS, with Alzheimer's disease (AD) considered as the "brain-type diabetes." In fact, brain insulin has been shown to regulate both peripheral and central glucose metabolism, neurotransmission, learning, and memory and to be neuroprotective. And a future challenge will be to unravel the complex interactions between aging and diabetes, which, we believe, will allow the development of efficient preventive and therapeutic strategies to overcome age-related diseases and to prolong human "healthy" longevity. Herewith, we aim to integrate the metabolic, neuromodulatory, and neuroprotective roles of insulin in two age-related pathologies: diabetes and AD, both in terms of intracellular signaling and potential therapeutic approach.
Collapse
Affiliation(s)
- Ana I. Duarte
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Paula I. Moreira
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
| | - Catarina R. Oliveira
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Institute of Biochemistry, Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
| |
Collapse
|
29
|
Masternak MM, Bartke A, Wang F, Spong A, Gesing A, Fang Y, Salmon AB, Hughes LF, Liberati T, Boparai R, Kopchick JJ, Westbrook R. Metabolic effects of intra-abdominal fat in GHRKO mice. Aging Cell 2012; 11:73-81. [PMID: 22040032 DOI: 10.1111/j.1474-9726.2011.00763.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Mice with targeted deletion of the growth hormone receptor (GHRKO mice) are growth hormone (GH) resistant, small, obese, hypoinsulinemic, highly insulin sensitive and remarkably long-lived. To elucidate the unexpected coexistence of adiposity with improved insulin sensitivity and extended longevity, we examined effects of surgical removal of visceral (epididymal and perinephric) fat on metabolic traits related to insulin signaling and longevity. Comparison of results obtained in GHRKO mice and in normal animals from the same strain revealed disparate effects of visceral fat removal (VFR) on insulin and glucose tolerance, adiponectin levels, accumulation of ectopic fat, phosphorylation of insulin signaling intermediates, body temperature, and respiratory quotient (RQ). Overall, VFR produced the expected improvements in insulin sensitivity and reduced body temperature and RQ in normal mice and had opposite effects in GHRKO mice. Some of the examined parameters were altered by VFR in opposite directions in GHRKO and normal mice, and others were affected in only one genotype or exhibited significant genotype × treatment interactions. Functional differences between visceral fat of GHRKO and normal mice were confirmed by measurements of adipokine secretion, lipolysis, and expression of genes related to fat metabolism. We conclude that in the absence of GH signaling, the secretory activity of visceral fat is profoundly altered and unexpectedly promotes enhanced insulin sensitivity. The apparent beneficial effects of visceral fat in GHRKO mice may also explain why reducing adiposity by calorie restriction fails to improve insulin signaling or further extend longevity in these animals.
Collapse
Affiliation(s)
- Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
List EO, Sackmann-Sala L, Berryman DE, Funk K, Kelder B, Gosney ES, Okada S, Ding J, Cruz-Topete D, Kopchick JJ. Endocrine parameters and phenotypes of the growth hormone receptor gene disrupted (GHR-/-) mouse. Endocr Rev 2011; 32:356-86. [PMID: 21123740 PMCID: PMC3365798 DOI: 10.1210/er.2010-0009] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Disruption of the GH receptor (GHR) gene eliminates GH-induced intracellular signaling and, thus, its biological actions. Therefore, the GHR gene disrupted mouse (GHR-/-) has been and is a valuable tool for helping to define various parameters of GH physiology. Since its creation in 1995, this mouse strain has been used by our laboratory and others for numerous studies ranging from growth to aging. Some of the most notable discoveries are their extreme insulin sensitivity in the presence of obesity. Also, the animals have an extended lifespan, which has generated a large number of investigations into the roles of GH and IGF-I in the aging process. This review summarizes the many results derived from the GHR-/- mice. We have attempted to present the findings in the context of current knowledge regarding GH action and, where applicable, to discuss how these mice compare to GH insensitivity syndrome in humans.
Collapse
Affiliation(s)
- Edward O List
- The Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Gesing A, Masternak MM, Wang F, Lewinski A, Karbownik-Lewinska M, Bartke A. Decreased expression level of apoptosis-related genes and/or proteins in skeletal muscles, but not in hearts, of growth hormone receptor knockout mice. Exp Biol Med (Maywood) 2011; 236:156-68. [PMID: 21321312 DOI: 10.1258/ebm.2010.010202] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The long-lived growth hormone (GH) receptor knockout (GHRKO; KO) mice are GH-resistant due to targeted disruption of the GH receptor (Ghr) gene. Apoptosis is a physiological process in which cells play an active role in their own death and is a normal component of the development and health of multicellular organisms. Aging is associated with the progressive loss of strength of skeletal and heart muscles. Calorie restriction (CR) is a well-known experimental model to delay aging and increase lifespan. The aim of the study was to examine the expression of the following apoptosis-related genes: caspase-3, caspase-9, caspase-8, bax, bcl-2, Smac/DIABLO, p53 and cytochrome c1 (cyc1) in the skeletal muscles and hearts of female normal and GHRKO mice, fed ad libitum or subjected to 40% CR for six months, starting at two months of age. Moreover, skeletal muscle caspase-3, caspase-9, caspase-8, bax, bcl-2, Smac/DIABLO, Apaf-1, bad, phospho-bad (pbad), phospho-p53 and cytochrome c (cyc) protein expression levels were assessed. Expression of caspase-3, caspase-9, bax and Smac/DIABLO genes and proteins was decreased in GHRKO's skeletal muscles. The Apaf-1 protein expression also was diminished in this tissue. In contrast, bcl-2 and pbad protein levels were increased in skeletal muscles in knockouts. No changes were demonstrated for the examined genes' expression in GHRKO's hearts except for the increased level of cyc1 mRNA. CR did not alter the expression of the examined genes and proteins in skeletal muscles of knockouts versus normal (N) mice. In heart homogenates, CR increased caspase-3 mRNA level as compared with ad libitum mice. Decreased expression of certain proapoptotic genes and/or proteins may constitute the potential mechanism of prolonged longevity in GHRKO mice, protecting these animals from aging; this potential beneficial mechanism is not affected by CR.
Collapse
Affiliation(s)
- Adam Gesing
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, 801 N Rutledge St., Room 4389, 62702-4910 Springfield, IL, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Chistyakova OV. Signaling pathway of insulin and insulin-like growth factor 1 (IGF-1) as a potential regulator of lifespan. J EVOL BIOCHEM PHYS+ 2011. [DOI: 10.1134/s0022093008010015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
33
|
Brewer GJ. Epigenetic oxidative redox shift (EORS) theory of aging unifies the free radical and insulin signaling theories. Exp Gerontol 2009; 45:173-9. [PMID: 19945522 DOI: 10.1016/j.exger.2009.11.007] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 11/17/2009] [Accepted: 11/18/2009] [Indexed: 12/14/2022]
Abstract
Harman's free radical theory of aging posits that oxidized macromolecules accumulate with age to decrease function and shorten life-span. However, nutritional and genetic interventions to boost anti-oxidants have generally failed to increase life-span. Furthermore, the free radical theory fails to explain why exercise causes higher levels of oxyradical damage, but generally promotes healthy aging. The separate anti-aging paradigms of genetic or caloric reductions in the insulin signaling pathway is thought to slow the rate of living to reduce metabolism, but recent evidence from Westbrook and Bartke suggests metabolism actually increases in long-lived mice. To unify these disparate theories and data, here, we propose the epigenetic oxidative redox shift (EORS) theory of aging. According to EORS, sedentary behavior associated with age triggers an oxidized redox shift and impaired mitochondrial function. In order to maintain resting energy levels, aerobic glycolysis is upregulated by redox-sensitive transcription factors. As emphasized by DeGrey, the need to supply NAD(+) for glucose oxidation and maintain redox balance with impaired mitochondrial NADH oxidoreductase requires the upregulation of other oxidoreductases. In contrast to the 2% inefficiency of mitochondrial reduction of oxygen to the oxyradical, these other oxidoreductases enable glycolytic energy production with a deleterious 100% efficiency in generating oxyradicals. To avoid this catastrophic cycle, lactate dehydrogenase is upregulated at the expense of lactic acid acidosis. This metabolic shift is epigenetically enforced, as is insulin resistance to reduce mitochondrial turnover. The low mitochondrial capacity for efficient production of energy reinforces a downward spiral of more sedentary behavior leading to accelerated aging, increased organ failure with stress, impaired immune and vascular functions and brain aging. Several steps in the pathway are amenable to reversal for exit from the vicious cycle of EORS. Examples from our work in the aging rodent brain as well as other aging models are provided.
Collapse
Affiliation(s)
- Gregory J Brewer
- Kenneth Stark Endowed Chair in Alzheimer Research, Southern Illinois University School of Medicine, Springfield, IL 62794-9626, USA.
| |
Collapse
|
34
|
Berryman DE, List EO, Palmer AJ, Chung MY, Wright-Piekarski J, Lubbers E, O'Connor P, Okada S, Kopchick JJ. Two-year body composition analyses of long-lived GHR null mice. J Gerontol A Biol Sci Med Sci 2009; 65:31-40. [PMID: 19901018 DOI: 10.1093/gerona/glp175] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Growth hormone receptor gene-disrupted (GHR-/-) mice exhibit increased life span and adipose tissue mass. Although this obese phenotype has been reported extensively for young adult male GHR-/- mice, data for females and for other ages in either gender are lacking. Thus, the purpose of this study was to evaluate body composition longitudinally in both male and female GHR-/- mice. Results show that GHR-/- mice have a greater percent fat mass with no significant difference in absolute fat mass throughout life. Lean mass shows an opposite trend with percent lean mass not significantly different between genotypes but absolute mass reduced in GHR-/- mice. Differences in body composition are more pronounced in male than in female mice, and both genders of GHR-/- mice show specific enlargement of the subcutaneous adipose depot. Along with previously published data, these results suggest a consistent and intriguing protective effect of excess fat mass in the subcutaneous region.
Collapse
Affiliation(s)
- Darlene E Berryman
- School of Human and Consumer Sciences, College of Health and Human Services, Ohio University, Athens, OH 45701, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Panici JA, Wang F, Bonkowski MS, Spong A, Bartke A, Pawlikowska L, Kwok PY, Masternak MM. Is altered expression of hepatic insulin-related genes in growth hormone receptor knockout mice due to GH resistance or a difference in biological life spans? J Gerontol A Biol Sci Med Sci 2009; 64:1126-33. [PMID: 19706698 DOI: 10.1093/gerona/glp111] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Growth hormone receptor knockout (GHRKO) mice live about 40%-55% longer than their normal (N) littermates. Previous studies of 21-month-old GHRKO and N mice showed major alterations of the hepatic expression of genes involved in insulin signaling. Differences detected at this age may have been caused by the knockout of the growth hormone receptor (GHR) or by differences in biological age between GHRKO and N mice. To address this question, we compared GHRKO and N mice at ages corresponding to the same percentage of median life span to see if the differences of gene expression persisted. Comparison of GHRKO and N mice at approximately 50% of biological life span showed significant differences in hepatic expression of all 14 analyzed genes. We conclude that these changes are due to disruption of GHR gene and the consequent suppression of growth hormone signaling rather than to differences in "biological age" between mutant and normal animals sampled at the same chronological age.
Collapse
Affiliation(s)
- Jacob A Panici
- Department of Internal Medicine, Geriatrics Research, Springfield, IL 62794-9628, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Garinis GA, Uittenboogaard LM, Stachelscheid H, Fousteri M, van Ijcken W, Breit TM, van Steeg H, Mullenders LH, van der Horst GT, Brüning JC, Niessen CM, Hoeijmakers JH, Schumacher B. Persistent transcription-blocking DNA lesions trigger somatic growth attenuation associated with longevity. Nat Cell Biol 2009; 11:604-15. [PMID: 19363488 PMCID: PMC2782455 DOI: 10.1038/ncb1866] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Accepted: 02/05/2009] [Indexed: 12/22/2022]
Abstract
The accumulation of stochastic DNA damage throughout an organism's lifespan is thought to contribute to ageing. Conversely, ageing seems to be phenotypically reproducible and regulated through genetic pathways such as the insulin-like growth factor-1 (IGF-1) and growth hormone (GH) receptors, which are central mediators of the somatic growth axis. Here we report that persistent DNA damage in primary cells from mice elicits changes in global gene expression similar to those occurring in various organs of naturally aged animals. We show that, as in ageing animals, the expression of IGF-1 receptor and GH receptor is attenuated, resulting in cellular resistance to IGF-1. This cell-autonomous attenuation is specifically induced by persistent lesions leading to stalling of RNA polymerase II in proliferating, quiescent and terminally differentiated cells; it is exacerbated and prolonged in cells from progeroid mice and confers resistance to oxidative stress. Our findings suggest that the accumulation of DNA damage in transcribed genes in most if not all tissues contributes to the ageing-associated shift from growth to somatic maintenance that triggers stress resistance and is thought to promote longevity.
Collapse
MESH Headings
- Aging/physiology
- Animal Structures/metabolism
- Animals
- DNA/radiation effects
- DNA Damage/physiology
- DNA Repair/physiology
- Gene Expression Profiling
- Growth/physiology
- Growth/radiation effects
- Humans
- Longevity/physiology
- Longevity/radiation effects
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Biological
- Neoplasms/etiology
- Neoplasms/genetics
- Oxidative Stress/physiology
- Progeria/genetics
- Progeria/metabolism
- RNA Polymerase II/metabolism
- Rats
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptors, Somatotropin/genetics
- Receptors, Somatotropin/metabolism
- Stress, Physiological/physiology
- Transcription, Genetic/genetics
- Transcription, Genetic/radiation effects
- Ultraviolet Rays
Collapse
Affiliation(s)
- George A. Garinis
- MGC Department of Cell Biology and Genetics, Center for Biomedical Genetics, Erasmus Medical Center, PO Box 1738, 3000 DR Rotterdam, The Netherlands
- Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| | - Lieneke M. Uittenboogaard
- MGC Department of Cell Biology and Genetics, Center for Biomedical Genetics, Erasmus Medical Center, PO Box 1738, 3000 DR Rotterdam, The Netherlands
| | - Heike Stachelscheid
- Center for Molecular Medicine Cologne, University of Cologne, Germany
- Institute for Genetics, University of Cologne, Germany
| | - Maria Fousteri
- Department of Toxicogenetics, LUMC, Leiden, The Netherlands
| | - Wilfred van Ijcken
- Erasmus Center for Biomics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Timo M. Breit
- Integrative Bioinformatics Unit, Institute for Informatics, Faculty of Science, University of Amsterdam, The Netherlands
| | - Harry van Steeg
- National Institute of Public Health and the Environment (RIVM), Laboratory of Toxicology, Pathology and Genetics (TOX), Bilthoven, The Netherlands
| | | | - Gijsbertus T.J. van der Horst
- MGC Department of Cell Biology and Genetics, Center for Biomedical Genetics, Erasmus Medical Center, PO Box 1738, 3000 DR Rotterdam, The Netherlands
| | - Jens C. Brüning
- Institute for Genetics, University of Cologne, Germany
- Cologne Excellence Cluster for Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany
| | - Carien M. Niessen
- Center for Molecular Medicine Cologne, University of Cologne, Germany
- Cologne Excellence Cluster for Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany
- Department of Dermatology, University of Cologne, Germany
| | - Jan H.J. Hoeijmakers
- MGC Department of Cell Biology and Genetics, Center for Biomedical Genetics, Erasmus Medical Center, PO Box 1738, 3000 DR Rotterdam, The Netherlands
| | - Björn Schumacher
- MGC Department of Cell Biology and Genetics, Center for Biomedical Genetics, Erasmus Medical Center, PO Box 1738, 3000 DR Rotterdam, The Netherlands
- Cologne Excellence Cluster for Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany
| |
Collapse
|
37
|
Chen JH, Martin-Gronert MS, Tarry-Adkins J, Ozanne SE. Maternal protein restriction affects postnatal growth and the expression of key proteins involved in lifespan regulation in mice. PLoS One 2009; 4:e4950. [PMID: 19308256 PMCID: PMC2654922 DOI: 10.1371/journal.pone.0004950] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Accepted: 02/25/2009] [Indexed: 01/21/2023] Open
Abstract
We previously reported that maternal protein restriction in rodents influenced the rate of growth in early life and ultimately affected longevity. Low birth weight caused by maternal protein restriction followed by catch-up growth (recuperated animals) was associated with shortened lifespan whereas protein restriction and slow growth during lactation (postnatal low protein: PLP animals) increased lifespan. We aim to explore the mechanistic basis by which these differences arise. Here we investigated effects of maternal diet on organ growth, metabolic parameters and the expression of insulin/IGF1 signalling proteins and Sirt1 in muscle of male mice at weaning. PLP mice which experienced protein restriction during lactation had lower fasting glucose (P = 0.038) and insulin levels (P = 0.046) suggesting improved insulin sensitivity. PLP mice had higher relative weights (adjusted by body weight) of brain (P = 0.0002) and thymus (P = 0.031) compared to controls suggesting that enhanced functional capacity of these two tissues is beneficial to longevity. They also had increased expression of insulin receptor substrate 1 (P = 0.021) and protein kinase C zeta (P = 0.046). Recuperated animals expressed decreased levels of many insulin signalling proteins including PI3 kinase subunits p85alpha (P = 0.018), p110beta (P = 0.048) and protein kinase C zeta (P = 0.006) which may predispose these animals to insulin resistance. Sirt1 protein expression was reduced in recuperated offspring. These observations suggest that maternal protein restriction can affect major metabolic pathways implicated in regulation of lifespan at a young age which may explain the impact of maternal diet on longevity.
Collapse
Affiliation(s)
- Jian-Hua Chen
- University of Cambridge Metabolic Research Laboratories Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, United Kingdom.
| | | | | | | |
Collapse
|
38
|
Berryman DE, Christiansen JS, Johannsson G, Thorner MO, Kopchick JJ. Role of the GH/IGF-1 axis in lifespan and healthspan: lessons from animal models. Growth Horm IGF Res 2008; 18:455-71. [PMID: 18710818 PMCID: PMC2631405 DOI: 10.1016/j.ghir.2008.05.005] [Citation(s) in RCA: 203] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Accepted: 05/02/2008] [Indexed: 12/18/2022]
Abstract
Animal models are fundamentally important in our quest to understand the genetic, epigenetic, and environmental factors that contribute to human aging. In comparison to humans, relatively short-lived mammals are useful models as they allow for rapid assessment of both genetic manipulation and environmental intervention as related to longevity. These models also allow for the study of clinically relevant pathologies as a function of aging. Data associated with more distant species offers additional insight and critical consideration of the basic physiological processes and molecular mechanisms that influence lifespan. Consistently, two interventions, caloric restriction and repression of the growth hormone (GH)/insulin-like growth factor-1/insulin axis, have been shown to increase lifespan in both invertebrates and vertebrate animal model systems. Caloric restriction (CR) is a nutrition intervention that robustly extends lifespan whether it is started early or later in life. Likewise, genes involved in the GH/IGF-1 signaling pathways can lengthen lifespan in vertebrates and invertebrates, implying evolutionary conservation of the molecular mechanisms. Specifically, insulin and insulin-like growth factor-1 (IGF-1)-like signaling and its downstream intracellular signaling molecules have been shown to be associated with lifespan in fruit flies and nematodes. More recently, mammalian models with reduced growth hormone (GH) and/or IGF-1 signaling have also been shown to have extended lifespans as compared to control siblings. Importantly, this research has also shown that these genetic alterations can keep the animals healthy and disease-free for longer periods and can alleviate specific age-related pathologies similar to what is observed for CR individuals. Thus, these mutations may not only extend lifespan but may also improve healthspan, the general health and quality of life of an organism as it ages. In this review, we will provide an overview of how the manipulation of the GH/IGF axis influences lifespan, highlight the invertebrate and vertebrate animal models with altered lifespan due to modifications to the GH/IGF-1 signaling cascade or homologous pathways, and discuss the basic phenotypic characteristics and healthspan of these models.
Collapse
Affiliation(s)
- Darlene E. Berryman
- School of Human and Consumer Sciences, College of Health and Human Services, Ohio University, Athens, OH 45701
| | - Jens Sandahl Christiansen
- Jens Sandahl Christiansen, Department of Endocrinology, Aarhus University Hospital, Kommunehospitalet, DK 8000 Aarhus, Denmark
| | - Gudmundur Johannsson
- Gudmundur Johannsson, MD, Research Centre for Endocrinology and Metabolism, Sahlgrenska University Hospital, S-413 45 Göteborg, Sweden
| | - Michael O. Thorner
- Michael O. Thorner, University of Virginia Health System, Endocrinology and Metabolism, Charlottesville, VA 22908
| | - John J. Kopchick
- Edison Biotechnology Institute and Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, Athens, OH 45701; Phone: (740)593-4534; Fax: (740)593-4795
| |
Collapse
|
39
|
Conover CA, Mason MA, Levine JA, Novak CM. Metabolic consequences of pregnancy-associated plasma protein-A deficiency in mice: exploring possible relationship to the longevity phenotype. J Endocrinol 2008; 198:599-605. [PMID: 18566100 PMCID: PMC2593875 DOI: 10.1677/joe-08-0179] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Mice born with the deletion of the gene for pregnancy-associated plasma protein-A (PAPP-A), a model of reduced local IGF activity, live approximately 30% longer than their wild-type (WT) littermates. In this study, we investigated metabolic consequences of PAPP-A gene deletion and possible relationship to lifespan extension. Specifically, we determined whether 18-month-old PAPP-A knockout (KO) mice when compared with their WT littermates have reduced energy expenditure and/or altered glucose-insulin sensitivity. Food intake, and total energy expenditure and resting energy expenditure as measured by calorimetry were not different between PAPP-A KO and WT mice when subjected to the analysis of covariance with body weight as the covariate. However, there was an increase in spontaneous physical activity in PAPP-A KO mice. Both WT and PAPP-A KO mice exhibited mild insulin resistance with age, as assessed by fasting glucose/insulin ratios. Oral glucose tolerance and insulin sensitivity were not significantly different between the two groups of mice, although there appeared to be a decrease in the average size of the pancreatic islets in PAPP-A KO mice. Thus, neither reduced 'rate of living' nor altered glucose-insulin homeostasis can be considered key determinants of the enhanced longevity of PAPP-A KO mice. These findings are discussed in the context of those from other long-lived mouse models.
Collapse
Affiliation(s)
- Cheryl A Conover
- Division of Endocrinology, Metabolism and Nutrition, Mayo Clinic, 200 First Street SW, 5-194 Joseph, Rochester, Minnesota 55905, USA.
| | | | | | | |
Collapse
|
40
|
Giani JF, Bonkowski MS, Munoz MC, Masternak MM, Turyn D, Bartke A, Dominici FP. Insulin Signaling Cascade in the Hearts of Long-Lived Growth Hormone Receptor Knockout Mice: Effects of Calorie Restriction. J Gerontol A Biol Sci Med Sci 2008; 63:788-97. [DOI: 10.1093/gerona/63.8.788] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
41
|
Chandrashekar V, Dawson CR, Martin ER, Rocha JS, Bartke A, Kopchick JJ. Age-related alterations in pituitary and testicular functions in long-lived growth hormone receptor gene-disrupted mice. Endocrinology 2007; 148:6019-25. [PMID: 17872367 DOI: 10.1210/en.2007-0837] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The somatotropic axis, GH, and IGF-I interact with the hypothalamic-pituitary-gonadal axis in health and disease. GH-resistant GH receptor-disrupted knockout (GHRKO) male mice are fertile but exhibit delayed puberty and decreases in plasma FSH levels, testicular content of LH, and prolactin (PRL) receptors, whereas PRL levels are elevated. Because the lifespan of GHRKO mice is much greater than the lifespan of their normal siblings, it was of interest to compare age-related changes in the hypothalamic-pituitary-gonadal axis in GHRKO and normal animals. Plasma IGF-I, insulin, PRL, LH, FSH, androstenedione and testosterone levels, and acute responses to GnRH and LH were measured in young (2-4 and 5-6 months of age) and old (18-19 and 23-26 months of age) male GHRKO mice and their normal siblings. Plasma IGF-I was not detectable in GHRKO mice. Plasma PRL levels increased with age in normal mice but declined in GHRKO males, and did not differ in old GHRKO and normal animals. Plasma LH responses to acute GnRH stimulation were attenuated in GHRKO mice but increased with age only in normal mice. Plasma FSH levels were decreased in GHRKO mice regardless of age. Plasma testosterone responses to LH stimulation were attenuated in old mice regardless of genotype, whereas plasma androstenedione responses were reduced with age only in GHRKO mice. Testicular IGF-I mRNA levels were normal in young and increased in old GHRKO mice, whereas testicular concentrations and total IGF-I levels were decreased in these animals. These findings indicate that GH resistance due to targeted disruption of the GH receptor gene in mice leads to suppression of testicular IGF-I levels, and modifies the effects of aging on plasma PRL levels and responses of the pituitary and testes to GnRH and LH stimulation. Plasma testosterone levels declined during aging in normal but not in GHRKO mice, and the age-related increase in the LH responses to exogenous GnRH was absent in GHRKO mice, perhaps reflecting a delay of aging in these remarkably long-lived animals.
Collapse
Affiliation(s)
- Varadaraj Chandrashekar
- Department Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA
| | | | | | | | | | | |
Collapse
|
42
|
Al-Regaiey KA, Masternak MM, Bonkowski MS, Panici JA, Kopchick JJ, Bartke A. Effects of caloric restriction and growth hormone resistance on insulin-related intermediates in the skeletal muscle. J Gerontol A Biol Sci Med Sci 2007; 62:18-26. [PMID: 17301033 DOI: 10.1093/gerona/62.1.18] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Growth hormone receptor-deficient (GHRKO) mice are long-lived and have reduced insulin-like growth factor (IGF)-1 and insulin levels and enhanced insulin sensitivity thus resembling the phenotype of animals subjected to calorie restriction (CR). In contrast to its effects in normal mice, CR does not improve insulin sensitivity or increase longevity in GHRKO males. In an attempt to identify mechanisms underlying this differential response to CR, effects of CR on the expression of insulin-related genes were compared in GHRKO and normal mice. In addition to changes detected in both genotypes, and responses unique to GHRKO mice, the levels of Akt2 and peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC1alpha) were increased and levels of phosphorylated c-Jun N-terminal kinase (JNK)1 were reduced in response to CR only in normal mice. These changes may be related to mechanisms of improving insulin sensitivity and life expectancy.
Collapse
Affiliation(s)
- Khalid A Al-Regaiey
- Departments of Internal Medicine, Southern Illinois University, School of Medicine, Springfield, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Leclercq IA, Lebrun VA, Stärkel P, Horsmans YJ. Intrahepatic insulin resistance in a murine model of steatohepatitis: effect of PPARgamma agonist pioglitazone. J Transl Med 2007; 87:56-65. [PMID: 17075577 DOI: 10.1038/labinvest.3700489] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hepatic insulin resistance is associated with hepatic steatosis and is thought to play an important role in the pathogenesis of steatohepatitis. Using a murine model of steatohepatitis (mice fed a diet deficient in methionine and choline-MCD diet), we tested the effects of the insulin-sensitising, PPARgamma agonist drug pioglitazone (PGZ) on systemic and intrahepatic insulin sensitivity and on liver pathology. Compared to controls, mice fed the MCD diet develop a significant steatohepatitis, have enhanced glucose tolerance and enhanced systemic response to insulin. PGZ did not affect the systemic insulin sensitivity in control or MCD-fed mice as assessed in vivo by intraperitoneal glucose or insulin dynamic tests. However, PGZ prevented hepatic fat accumulation and steatohepatitis induced by the MCD diet. This effect was associated with an increased mass of adipose tissue and increased expression and release of adiponectin, while hepatic acyl co-enzyme A oxidase and acyl-co-enzyme A carboxylase, regulating hepatic beta-oxidation of fatty acid, remained unchanged. Steatohepatitis in MCD-diet-fed mice was associated with intrahepatic insulin resistance as shown by a reduced phosphorylation of hepatic insulin receptor, and Akt in response to an insulin stimulus. PGZ to MCD-fed mice restored the activation of the insulin receptor and of the Akt pathway in response to insulin. In conclusion, PGZ alleviates steatosis and steatohepatitis induced by the MCD diet, an effect associated with correction of intrahepatic insulin resistance.
Collapse
Affiliation(s)
- Isabelle A Leclercq
- Laboratory of Gastroenterology, Faculty of Medicine, Université Catholique de Louvain (UCL), Brussels, Belgium.
| | | | | | | |
Collapse
|
44
|
Abstract
Sustaining health and extending longevity have been perpetual goals of all human societies. For almost as long, there has been an ongoing effort to develop treatments that could prevent aging and, more importantly, make us live longer and more healthily. At present, there is one known intervention that delays aging, increases lifespan and prevents diseases in many animal species: calorie restriction. There are other physiological factors that are believed to have corresponding impacts on longevity and aging, including growth hormone and the insulin/insulin-like growth factor 1 signaling pathway. However, there is still much debate regarding the complex action of growth hormone on lifespan and aging.
Collapse
Affiliation(s)
- Michal M Masternak
- a Southern Illinois University, School of Medicine, Geriatrics Research, Department of Internal Medicine, 801 N. Rutledge Street, Room 4389, PO Box 19628, Springfield, IL 62794-9628, USA.
| | - Andrzej Bartke
- b Southern Illinois University, School of Medicine, Geriatrics Research, Department of Internal Medicine and Physiology, 801 N. Rutledge Street, Room 4389, PO Box 19628, Springfield, IL 62794-9628, USA.
| |
Collapse
|
45
|
Coschigano KT. Aging-related characteristics of growth hormone receptor/binding protein gene-disrupted mice. AGE (DORDRECHT, NETHERLANDS) 2006; 28:191-200. [PMID: 19943140 PMCID: PMC2464722 DOI: 10.1007/s11357-006-9004-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Accepted: 12/01/2005] [Indexed: 05/28/2023]
Abstract
Since generation of the growth hormone receptor/binding protein (GHR/BP) gene-disrupted mouse nearly 10 years ago, use of this mouse model has become widespread in the elucidation of the physiological roles of GH and insulin-like growth factor-1 (IGF-1). In particular, it serves as a useful model to study mechanisms of aging. This review highlights the evidence demonstrating that the loss of GH signaling leads to lifespan extension in mice, and presents the multiple characteristics of this mouse line that suggest the life extension is due to alteration of the aging process.
Collapse
Affiliation(s)
- Karen T Coschigano
- Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, 351 Irvine Hall, Athens, OH 45701, USA.
| |
Collapse
|
46
|
Bonkowski MS, Rocha JS, Masternak MM, Al Regaiey KA, Bartke A. Targeted disruption of growth hormone receptor interferes with the beneficial actions of calorie restriction. Proc Natl Acad Sci U S A 2006; 103:7901-5. [PMID: 16682650 PMCID: PMC1458512 DOI: 10.1073/pnas.0600161103] [Citation(s) in RCA: 235] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Reduced intake of nutrients [calorie restriction (CR)] extends longevity in organisms ranging from yeast to mammals. Mutations affecting somatotropic, insulin, or homologous signaling pathways can increase life span in worms, flies, and mice, and there is considerable evidence that reduced secretion of insulin-like growth factor I and insulin are among the mechanisms that mediate the effects of CR on aging and longevity in mammals. In the present study, mice with targeted disruption of the growth hormone (GH) receptor [GH receptor/GH-binding protein knockout (GHRKO) mice] and their normal siblings were fed ad libitum (AL) or subjected to 30% CR starting at 2 months of age. In normal females and males, CR produced the expected increases in overall, average, median, and maximal life span. Longevity of normal mice subjected to CR resembles that of GHRKO animals fed AL. In sharp contrast to its effects in normal mice, CR failed to increase overall, median, or average life span in GHRKO mice and increased maximal life span only in females. In a separate group of animals, CR for 1 year improved insulin sensitivity in normal mice but failed to further enhance the remarkable insulin sensitivity in GHRKO mutants. These data imply that somatotropic signaling is critically important not only in the control of aging and longevity under conditions of unlimited food supply but also in mediating the effects of CR on life span. The present findings also support the notion that enhanced sensitivity to insulin plays a prominent role in the actions of CR and GH resistance on longevity.
Collapse
Affiliation(s)
| | | | | | | | - Andrzej Bartke
- Departments of *Internal Medicine–Geriatrics Research
- Physiology, Southern Illinois University School of Medicine, Springfield, IL 62794
- To whom correspondence should be addressed at:
Geriatrics Research, Southern Illinois University, 801 N. Rutledge, P.O. Box 19628, Springfield, IL 62794-9628. E-mail:
| |
Collapse
|
47
|
Ingram DK, Zhu M, Mamczarz J, Zou S, Lane MA, Roth GS, deCabo R. Calorie restriction mimetics: an emerging research field. Aging Cell 2006; 5:97-108. [PMID: 16626389 DOI: 10.1111/j.1474-9726.2006.00202.x] [Citation(s) in RCA: 292] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
When considering all possible aging interventions evaluated to date, it is clear that calorie restriction (CR) remains the most robust. Studies in numerous species have demonstrated that reduction of calories 30-50% below ad libitum levels of a nutritious diet can increase lifespan, reduce the incidence and delay the onset of age-related diseases, improve stress resistance, and decelerate functional decline. A current major focus of this research area is whether this nutritional intervention is relevant to human aging. Evidence emerging from studies in rhesus monkeys suggests that their response to CR parallels that observed in rodents. To assess CR effects in humans, clinical trials have been initiated. However, even if results from these studies could eventually substantiate CR as an effective pro-longevity strategy for humans, the utility of this intervention would be hampered because of the degree and length of restriction required. As an alternative strategy, new research has focused on the development of 'CR mimetics'. The objective of this strategy is to identify compounds that mimic CR effects by targeting metabolic and stress response pathways affected by CR, but without actually restricting caloric intake. For example, drugs that inhibit glycolysis (2-deoxyglucose), enhance insulin action (metformin), or affect stress signaling pathways (resveratrol), are being assessed as CR mimetics (CRM). Promising results have emerged from initial studies regarding physiological responses which resemble those observed in CR (e.g. reduced body temperature and plasma insulin) as well as protection against neurotoxicity (e.g. enhanced dopamine action and up-regulated neurotrophic factors). Ultimately, lifespan analyses in addition to expanded toxicity studies must be accomplished to fully assess the potential of any CRM. Nonetheless, this strategy clearly offers a very promising and expanding research endeavor.
Collapse
Affiliation(s)
- Donald K Ingram
- Laboratory of Experimental Gerontology, Intramural Research Program, National Institute on Aging, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Masternak MM, Al-Regaiey KA, Del Rosario Lim MM, Jimenez-Ortega V, Panici JA, Bonkowski MS, Kopchick JJ, Wang Z, Bartke A. Caloric restriction and growth hormone receptor knockout: effects on expression of genes involved in insulin action in the heart. Exp Gerontol 2006; 41:417-29. [PMID: 16524678 PMCID: PMC3082456 DOI: 10.1016/j.exger.2006.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2005] [Revised: 01/20/2006] [Accepted: 01/24/2006] [Indexed: 01/16/2023]
Abstract
Blockade of growth hormone (GH), decreased insulin-like growth factor-1 (IGF1) action and increased insulin sensitivity are associated with life extension and an apparent slowing of the aging process. We examined expression of genes involved in insulin action, IR, IRS1, IRS2, IGF1, IGF1R, GLUT4, PPARs and RXRs in the hearts of normal and GHR-/- (KO) mice fed ad libitum or subjected to 30% caloric restriction (CR). CR increased the cardiac expression of IR, IRS1, IGF1, IGF1R and GLUT4 in normal mice and IRS1, GLUT4, PPARalpha and PPARbeta/delta in GHR-KO animals. Expression of IR, IRS1, IRS2, IGF1, GLUT4, PPARgamma and PPARalpha did not differ between GHR-KO and normal mice. These unexpected results suggest that CR may lead to major modifications of insulin action in the heart, but high insulin sensitivity of GHR-KO mice is not associated with alterations in the levels of most of the examined molecules related to intracellular insulin signaling.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Blotting, Western
- Caloric Restriction
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Gene Expression
- Glucose Transporter Type 4/genetics
- Glucose Transporter Type 4/metabolism
- Growth Hormone/genetics
- Growth Hormone/metabolism
- Insulin/metabolism
- Insulin Resistance
- Insulin-Like Growth Factor I/genetics
- Insulin-Like Growth Factor I/metabolism
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Myocardium/metabolism
- PPAR alpha/genetics
- PPAR alpha/metabolism
- PPAR delta/genetics
- PPAR delta/metabolism
- PPAR gamma/genetics
- PPAR gamma/metabolism
- PPAR-beta/genetics
- PPAR-beta/metabolism
- RNA, Messenger/analysis
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Receptors, Somatotropin/genetics
- Receptors, Somatotropin/metabolism
- Retinoid X Receptors/genetics
- Retinoid X Receptors/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Michal M Masternak
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|