1
|
Qi Y, Wang X, Zhu C, Mi B, Cui C, Chen S, Zhao Z, Zhao F, Liu X, Wang J, Shi B, Hu J. Mutations in the FOXO3 Gene and Their Effects on Meat Traits in Gannan Yaks. Int J Mol Sci 2024; 25:1948. [PMID: 38396627 PMCID: PMC10888663 DOI: 10.3390/ijms25041948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The FOXO3 gene, a prominent member of the FOXO family, has been identified as a potential quantitative trait locus for muscle atrophy and lipid metabolism in livestock. It is also considered a promising candidate gene for meat quality traits such as Warner-Bratzler shear force (WBSF) and water holding capacity (WHC). The aim of this study was to identify sequence mutations in the FOXO3 gene of yaks and to analyze the association of genotypes and haplotypes with meat traits such as WBSF and WHC. Quantitative reverse-transcriptase PCR (RT-qPCR) was applied to determine the expression levels of FOXO3 in yak tissues, with the results revealing a high expression in the yak longissimus dorsi muscle. Exons of the FOXO3 gene were then sequenced in 572 yaks using hybrid pool sequencing. Five single nucleotide polymorphisms were identified. Additionally, four effective haplotypes and four combined haplotypes were constructed. Two mutations of the FOXO3 gene, namely C>G at exon g.636 and A>G at exon g.1296, were associated with cooked meat percentage (CMP) (p < 0.05) and WBSF (p < 0.05), respectively. Furthermore, the WBSF of the H2H3 haplotype combination was significantly lower than that of other combinations (p < 0.05). The findings of this study suggest that genetic variations in FOXO3 could be a promising biomarker for improving yak meat traits.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Bingang Shi
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.Q.); (X.W.); (C.Z.); (B.M.); (C.C.); (S.C.); (Z.Z.); (F.Z.); (X.L.); (J.W.)
| | - Jiang Hu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.Q.); (X.W.); (C.Z.); (B.M.); (C.C.); (S.C.); (Z.Z.); (F.Z.); (X.L.); (J.W.)
| |
Collapse
|
2
|
Santos BF, Grenho I, Martel PJ, Ferreira BI, Link W. FOXO family isoforms. Cell Death Dis 2023; 14:702. [PMID: 37891184 PMCID: PMC10611805 DOI: 10.1038/s41419-023-06177-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/30/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023]
Abstract
FOXO family of proteins are transcription factors involved in many physiological and pathological processes including cellular homeostasis, stem cell maintenance, cancer, metabolic, and cardiovascular diseases. Genetic evidence has been accumulating to suggest a prominent role of FOXOs in lifespan regulation in animal systems from hydra, C elegans, Drosophila, and mice. Together with the observation that FOXO3 is the second most replicated gene associated with extreme human longevity suggests that pharmacological targeting of FOXO proteins can be a promising approach to treat cancer and other age-related diseases and extend life and health span. However, due to the broad range of cellular functions of the FOXO family members FOXO1, 3, 4, and 6, isoform-specific targeting of FOXOs might lead to greater benefits and cause fewer side effects. Therefore, a deeper understanding of the common and specific features of these proteins as well as their redundant and specific functions in our cells represents the basis of specific targeting strategies. In this review, we provide an overview of the evolution, structure, function, and disease-relevance of each of the FOXO family members.
Collapse
Affiliation(s)
- Bruno F Santos
- Algarve Biomedical Center Research Institute-ABC-RI, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
- Centro Hospitalar Universitário do Algarve (CHUA). Rua Leão Penedo, 8000-386, Faro, Portugal
| | - Inês Grenho
- Algarve Biomedical Center Research Institute-ABC-RI, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
| | - Paulo J Martel
- Center for Health Technology and Services Research (CINTESIS)@RISE, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
| | - Bibiana I Ferreira
- Algarve Biomedical Center Research Institute-ABC-RI, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
- Algarve Biomedical Center (ABC), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.
| | - Wolfgang Link
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM). Arturo Duperier 4, 28029, Madrid, Spain.
| |
Collapse
|
3
|
Leszczyńska D, Szatko A, Papierska L, Zgliczyński W, Glinicki P. Musculoskeletal complications of Cushing syndrome. Reumatologia 2023; 61:271-282. [PMID: 37745145 PMCID: PMC10515123 DOI: 10.5114/reum/169889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/22/2023] [Indexed: 09/26/2023] Open
Abstract
Prolonged exposure to an excess of glucocorticosteroids (GCs), both endogenous and exogenous, leads to a wide range of comorbidities, including cardiovascular, metabolic, psychiatric, and musculoskeletal disorders. The latter comprise osteopenia and osteoporosis leading to skeletal fractures and myopathy. Although endogenous hypercortisolemia is a rare disorder, GCs are among the most frequently prescribed drugs, often administered chronically and despite multiple side effects, impossible to taper off due to therapeutic reasons. The pathophysiology of the effect of GC excess on bone often leads to fractures despite normal or low-normal bone mineral density and it includes direct (mainly disturbance in bone formation processes, through inactivation of the Wnt/β-catenin signalling pathway) and indirect mechanisms (through suppressing the gonadal and somatotrophic axis, and also through antagonizing vitamin D actions). Glucocorticosteroid-induced fast-twitch, glycolytic muscles atrophy occurs due to increased protein catabolism and impaired synthesis. Protein degradation is a result of activation of the ubiquitin proteasome and the lysosomes stimulated through overexpression of several atrogenes (such as FOXO-1 and atrogin-1). This review will discuss pathophysiology, clinical presentation, prevention, and management of GC-induced osteoporosis (including calcium and vitamin D supplementation, and bisphosphonates) and myopathy associated with GC excess.
Collapse
Affiliation(s)
- Dorota Leszczyńska
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, Warsaw, Poland
| | - Alicja Szatko
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, Warsaw, Poland
| | - Lucyna Papierska
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, Warsaw, Poland
| | - Wojciech Zgliczyński
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, Warsaw, Poland
| | - Piotr Glinicki
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, Warsaw, Poland
| |
Collapse
|
4
|
Hardiany NS, Remifta Putra MA, Penantian RM, Antarianto RD. Effects of fasting on FOXO3 expression as an anti-aging biomarker in the liver. Heliyon 2023; 9:e13144. [PMID: 36718153 PMCID: PMC9883274 DOI: 10.1016/j.heliyon.2023.e13144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 01/04/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Background Aging is a multifactorial degenerative process that can be modulated by fasting through activation of the Fork-head transcription factor of the O class 3 (FOXO3), which plays an important role in increasing lifespans. However, the effects of different fasting durations on the expression of FOXO3 in the liver has not yet been reported. Objective This study analyzed the effects of different fasting durations on the FOXO3 expression and its pathway by measuring sirtuin1 (SIRT1), insulin-like growth factor-1 (IGF-1), and superoxide dismutase (SOD) activity in the liver. Methods New Zealand white rabbits were used to mimic the effects of fasting on humans. The rabbits were divided into the control, intermittent fasting (IF), and prolonged fasting (PF) groups. Both fasting groups were interspersed with the non-fasting phase for 8 h. This treatment was conducted for 6 days. On Day 7, all the rabbits were sacrificed, and their livers were taken to measure the FOXO3 and SIRT1 mRNA expressions, the IGF-1 protein level, and the SOD activity level. ANOVA, multiple comparison, and Pearson's correlation were performed for statistical analysis. Results The FOXO3 and SIRT1 mRNA expressions were significantly higher in the IF group than in the control group. The FOXO3 expression was also 2.5 times higher in the IF group than in the PF group. There was a positive correlation between the FOXO3 and SIRT1 mRNA expressions. The IGF-1 protein level was significantly lower in the IF and PF groups than in the control group. The SOD-specific activity level was significantly higher in the IF group than in the control and PF groups. Conclusions Intermittent fasting significantly increased the FOXO3 and SIRT1 mRNA expressions and the SOD activity level in the livers of the rabbits and significantly decreased the circulating and hepatic IGF-1. Therefore, intermittent fasting may give a protective intervention effect towards aging.
Collapse
Affiliation(s)
- Novi Silvia Hardiany
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia,Corresponding author.
| | | | | | | |
Collapse
|
5
|
Tranilast protects pancreatic β-cells from palmitic acid-induced lipotoxicity via FoxO-1 inhibition. Sci Rep 2023; 13:101. [PMID: 36596838 PMCID: PMC9810694 DOI: 10.1038/s41598-022-25428-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/29/2022] [Indexed: 01/05/2023] Open
Abstract
Tranilast, an anti-allergic drug used in the treatment of bronchial asthma, was identified as an inhibitor of the transcription factor Forkhead box O-1 (FoxO-1) by high throughput chemical library screening in the present study. Based on FoxO-1's role in apoptotic cell death and differentiation, we examined the effect of tranilast on palmitic acid (PA)-induced cell damage in INS-1 cells. Tranilast substantially inhibited lipoapoptosis and restored glucose-stimulated insulin secretion under high PA exposure. Moreover, PA-mediated downregulation of PDX-1, MafA, and insulin expression was attenuated by tranilast. PA-induced oxidative and ER stress were also reduced in the presence of tranilast. These protective effects were accompanied by increased phosphorylation and decreased nuclear translocation of FoxO-1. Conversely, the effects of tranilast were diminished when treated in transfected cells with FoxO-1 phosphorylation mutant (S256A), suggesting that the tranilast-mediated effects are associated with inactivation of FoxO-1. Examination of the in vivo effects of tranilast using wild type and diabetic db/db mice showed improved glucose tolerance along with FoxO-1 inactivation in the pancreas of the tranilast-treated groups. Thus, we report here that tranilast has protective effects against PA-induced lipotoxic stress in INS-1 cells, at least partly, via FoxO-1 inactivation, which results in improved glucose tolerance in vivo.
Collapse
|
6
|
Ma K, Huang F, Qiao R, Miao L. Pathogenesis of sarcopenia in chronic obstructive pulmonary disease. Front Physiol 2022; 13:850964. [PMID: 35928562 PMCID: PMC9343800 DOI: 10.3389/fphys.2022.850964] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common pulmonary disease characterized by persistent respiratory symptoms and airflow obstruction. In addition to lung diseases, chronic obstructive pulmonary disease (COPD) is often associated with other organ diseases, and sarcopenia is one of the common diseases. In recent years, multiple factors have been proposed to influence muscle dysfunction in COPD patients, including systemic and local inflammation, oxidative stress, hypoxia, hypercapnia, protein synthesis, catabolic imbalance, nutritional changes, disuse, ageing, and the use of medications such as steroids. These factors alone or in combination can lead to a reduction in muscle mass and cross-sectional area, deterioration of muscle bioenergy metabolism, defects in muscle repair and regeneration mechanisms, apoptosis and other anatomical and/or functional pathological changes, resulting in a decrease in the muscle’s ability to work. This article reviews the research progress of possible pathogenesis of sarcopenia in COPD.
Collapse
|
7
|
Laskin GR, Gordon BS. The influence of nutrients on mechanical overload-induced changes to skeletal muscle mRNA content. Physiol Genomics 2022; 54:360-369. [PMID: 35848636 DOI: 10.1152/physiolgenomics.00075.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mechanical overload and nutrients influence skeletal muscle phenotype, with the combination sometimes having a synergistic effect. Muscle phenotypes influenced by these stimuli are mediated in part by changes to the muscle mRNA signature. However, the mechanical overload-sensitive gene programs that are influenced by nutrients remain unclear. The purpose of this study was to identify mechanical overload-sensitive gene programs that are influenced by nutrients and identify potential transcription factors that may differentiate the change in mRNA in response to mechanical overload versus nutrients. Nutrient deprived 12-week-old male mice were randomized to remain fasted or allowed access to food. All mice underwent a single bout of unilateral high force contractions of the tibialis anterior (TA). Four hours post-contractions TA muscles were extracted and content of 12 contraction-sensitive mRNAs were analyzed. The mRNA content of genes associated with Transcription, PI3K-Akt Signaling Pathway, Z-Disc, Intracellular Signal Transduction, Cell Cycle, and Amino Acid Transport was altered by contractions without influence of nutrient consumption. Conversely, the mRNA content of genes associated with Transcription, Cell Cycle, FoxO Signaling Pathway, and Amino Acid Transport was altered by contractions with nutrition consumption influencing the change. We identified Signal transducer and activator of transcription 3 (STAT3) and Activator protein 1 (AP-1) as transcription factors common amongst mRNAs that were primarily altered by mechanical overload regardless of feeding. Overall, these data provide a deeper molecular basis for the specific muscle phenotypes exclusive to mechanical overload versus those regulated by the addition of nutrients.
Collapse
Affiliation(s)
- Grant R Laskin
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, United States
| | - Bradley S Gordon
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, United States.,Institute of Sports Sciences and Medicine, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
8
|
MicroRNA-185 modulates CYP7A1 mediated cholesterol-bile acid metabolism through post-transcriptional and post-translational regulation of FoxO1. Atherosclerosis 2022; 348:56-67. [DOI: 10.1016/j.atherosclerosis.2022.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 12/22/2022]
|
9
|
Wang Y, Cheng Q, Su Q, Yu X, Shen T, Yang X, Jia W. Aesculin offers increased resistance against oxidative stress and protective effects against Aβ-induced neurotoxicity in Caenorhabditis elegans. Eur J Pharmacol 2022; 917:174755. [PMID: 35016885 DOI: 10.1016/j.ejphar.2022.174755] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/11/2021] [Accepted: 01/06/2022] [Indexed: 11/21/2022]
Abstract
Aesculin, a coumarin compound, is one of the major active ingredients of traditional Chinese herbal medicine Qinpi (Cortex Fraxini), which has been reported to exhibit antioxidative, anti-inflammatory and neuroprotective properties against oxidative stress and cellular apoptosis. However, the regulatory mechanisms remain poorly characterized in vivo. This research was performed to explore the underlying molecular mechanisms behind aesculin response conferring oxidative stress resistance, and the protective effects on amyloid-β (Aβ)-mediated neurotoxicity in Caenorhabditis elegans. Study indicated that aesculin plays the protective roles for C. elegans against oxidative stress and Aβ-mediated neurotoxicity and reduces the elevated ROS and MDA contents through enhancement of antioxidant defenses. The KEGG pathway analysis suggested that the differentially expressed genes are mainly involved in longevity regulating pathway, and the nuclear translocation of DAF-16 and the RNAi of daf-16 and hsf-1 indicated that DAF-16 and HSF-1 play critical roles in integrating upstream signals and inducing the expressions of stress resistance-related genes. Furthermore, the up-regulated expressions of their target genes such as sod-3 and hsp-16.2 were confirmed in transgenic GFP reporter strains CF1553 and CL2070, respectively. These results indicated that the regulators DAF-16 and HSF-1 elevate the stress resistance of C. elegans by modulating stress-responsive genes. Further experiments revealed that aesculin is capable of suppressing Aβ-induced oxidative stress and apoptosis and improves chemosensory behavior dysfunction in Aβ-transgenic nematodes. In summary, this study suggested that aesculin offers increased resistance against oxidative stress and protective effects against Aβ-induced neurotoxicity through activation of stress regulators DAF-16 and HSF-1 in nematodes.
Collapse
Affiliation(s)
- Ying Wang
- School of Biosciences & Biopharmaceutics, and Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510641, China
| | - Qiong Cheng
- School of Biosciences & Biopharmaceutics, and Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Qina Su
- School of Biosciences & Biopharmaceutics, and Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xuesong Yu
- School of Biosciences & Biopharmaceutics, and Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Tianqi Shen
- School of Biosciences & Biopharmaceutics, and Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xiaoling Yang
- School of Biosciences & Biopharmaceutics, and Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Weizhang Jia
- School of Biosciences & Biopharmaceutics, and Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
10
|
Zhang X, Luan P, Cao D, Hu G. A High-Density Genetic Linkage Map and Fine Mapping of QTL For Feed Conversion Efficiency in Common Carp ( Cyprinus carpio). Front Genet 2021; 12:778487. [PMID: 34868267 PMCID: PMC8633483 DOI: 10.3389/fgene.2021.778487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/22/2021] [Indexed: 12/02/2022] Open
Abstract
Feed conversion efficiency (FCE) is an economically crucial trait in fish, however, little progress has been made in genetics and genomics for this trait because phenotypes of the trait are difficult to measure. In this study, we constructed a high-density and high-resolution genetic linkage map with 28,416 SNP markers for common carp (Cyprinus carpio) based on high throughput genotyping with the carp 250K single nucleotide polymorphism (SNP) array in a full-sib F1 family of mirror carp (Cyprinus carpio) consisting of 141 progenies. The linkage map contained 11,983 distinct loci and spanned 3,590.09 cM with an average locus interval of 0.33 cM. A total of 17 QTL for the FCE trait were detected on four LGs (LG9, LG20, LG28, and LG32), explaining 8.9-15.9% of the phenotypic variations. One major cluster containing eight QTL (qFCE1-28, qFCE2-28, qFCE3-28, qFCE4-28, qFCE5-28, qFCE6-28, qFCE7-28, and qFCE8-28) was detected on LG28. Two clusters consisting of four QTL (qFCE1-32, qFCE2-32, qFCE3-32, and qFCE4-32) and three QTL (qFCE1-20, qFCE2-20, and qFCE3-20) were detected on LG32 and LG20, respectively. Nine candidate genes (ACACA, SCAF4, SLC2A5, TNMD, PCDH1, FOXO, AGO1, FFAR3, and ARID1A) underlying the feed efficiency trait were also identified, the biological functions of which may be involved in lipid metabolism, carbohydrate metabolism, energy deposition, fat accumulation, digestion, growth regulation, and cell proliferation and differentiation according to GO (Gene Ontology). As an important tool, high-density and high-resolution genetic linkage maps play a crucial role in the QTL fine mapping of economically important traits. Our novel findings provided new insights that elucidate the genetic basis and molecular mechanism of feed efficiency and the subsequent marker-assisted selection breeding in common carp.
Collapse
Affiliation(s)
- Xiaofeng Zhang
- National and Local United Engineering Laboratory for Freshwater Fish Breeding, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | | | | | - Guo Hu
- National and Local United Engineering Laboratory for Freshwater Fish Breeding, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| |
Collapse
|
11
|
Yang Z, Roth K, Agarwal M, Liu W, Petriello MC. The transcription factors CREBH, PPARa, and FOXO1 as critical hepatic mediators of diet-induced metabolic dysregulation. J Nutr Biochem 2021; 95:108633. [PMID: 33789150 PMCID: PMC8355060 DOI: 10.1016/j.jnutbio.2021.108633] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/31/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
The liver is a critical mediator of lipid and/or glucose homeostasis and is a primary organ involved in dynamic changes during feeding and fasting. Additionally, hepatic-centric pathways are prone to dysregulation during pathophysiological states including metabolic syndrome (MetS) and non-alcoholic fatty liver disease. Omics platforms and GWAS have elucidated genes related to increased risk of developing MetS and related disorders, but mutations in these metabolism-related genes are rare and cannot fully explain the increasing prevalence of MetS-related pathologies worldwide. Complex interactions between diet, lifestyle, environmental factors, and genetic predisposition jointly determine inter-individual variability of disease risk. Given the complexity of these interactions, researchers have focused on master regulators of metabolic responses incorporating and mediating the impact of multiple environmental cues. Transcription factors are DNA binding, terminal executors of signaling pathways that modulate the cellular responses to complex metabolic stimuli and are related to the control of hepatic lipid and glucose homeostasis. Among numerous hepatic transcription factors involved in regulating metabolism, three emerge as key players in transducing nutrient sensing, which are dysregulated in MetS-related perturbations in both clinical and preclinical studies: cAMP Responsive Element Binding Protein 3 Like 3 (CREB3L3), Peroxisome Proliferator Activated Receptor Alpha (PPAR), and Forkhead Box O1 (FOXO1). Additionally, these three transcription factors appear to be amenable to dietary and/or nutrient-based therapies, being potential targets of nutritional therapy. In this review we aim to describe the activation, regulation, and impact of these transcription factors in the context of metabolic homeostasis. We also summarize their perspectives in MetS and nutritional therapies.
Collapse
Affiliation(s)
- Zhao Yang
- Institute of Environmental Health Sciences (IEHS), Wayne State University, Detroit, MI, USA
| | - Katherine Roth
- Institute of Environmental Health Sciences (IEHS), Wayne State University, Detroit, MI, USA
| | - Manisha Agarwal
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Wanqing Liu
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA; Department of Pharmaceutical Sciences, College of Pharmacy, Wayne State University, Detroit, MI, USA
| | - Michael C Petriello
- Institute of Environmental Health Sciences (IEHS), Wayne State University, Detroit, MI, USA; Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
12
|
Zia A, Pourbagher-Shahri AM, Farkhondeh T, Samarghandian S. Molecular and cellular pathways contributing to brain aging. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2021; 17:6. [PMID: 34118939 PMCID: PMC8199306 DOI: 10.1186/s12993-021-00179-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022]
Abstract
Aging is the leading risk factor for several age-associated diseases such as neurodegenerative diseases. Understanding the biology of aging mechanisms is essential to the pursuit of brain health. In this regard, brain aging is defined by a gradual decrease in neurophysiological functions, impaired adaptive neuroplasticity, dysregulation of neuronal Ca2+ homeostasis, neuroinflammation, and oxidatively modified molecules and organelles. Numerous pathways lead to brain aging, including increased oxidative stress, inflammation, disturbances in energy metabolism such as deregulated autophagy, mitochondrial dysfunction, and IGF-1, mTOR, ROS, AMPK, SIRTs, and p53 as central modulators of the metabolic control, connecting aging to the pathways, which lead to neurodegenerative disorders. Also, calorie restriction (CR), physical exercise, and mental activities can extend lifespan and increase nervous system resistance to age-associated neurodegenerative diseases. The neuroprotective effect of CR involves increased protection against ROS generation, maintenance of cellular Ca2+ homeostasis, and inhibition of apoptosis. The recent evidence about the modem molecular and cellular methods in neurobiology to brain aging is exhibiting a significant potential in brain cells for adaptation to aging and resistance to neurodegenerative disorders.
Collapse
Affiliation(s)
- Aliabbas Zia
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ali Mohammad Pourbagher-Shahri
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), 9717853577 Birjand, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
13
|
Choi HE, Kim Y, Lee HJ, Cheon HG. Novel FoxO1 inhibitor, JY-2, ameliorates palmitic acid-induced lipotoxicity and gluconeogenesis in a murine model. Eur J Pharmacol 2021; 899:174011. [PMID: 33705803 DOI: 10.1016/j.ejphar.2021.174011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/26/2021] [Accepted: 02/28/2021] [Indexed: 11/25/2022]
Abstract
Forkhead transcription factor forkhead box O1 (FoxO1) plays an important role in glucose and lipid metabolism, contributing to the pathogenesis of metabolic disorders. This study aimed to discover a novel FoxO1 inhibitor as a potential new anti-diabetic drug candidate, and describes the biological effects of JY-2, 5-(2,4-dichlorophenyl)-3-(pyridin-2-yl)-1,2,4-oxadiazole in vitro and in vivo. JY-2 inhibited FoxO1 transcriptional activity in a concentration-dependent manner, with an IC50 value of 22 μM. The inhibitory effects of JY-2 on FoxO3a and FoxO4 appeared to be weaker than that on FoxO1. Consistent with its inhibitory effect on FoxO1, JY-2 reduced the palmitic acid (PA)-stimulated mRNA expression of glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK), two key enzymes involved in gluconeogenesis in HepG2 cells. In association with the reduced expression of lipid metabolism genes, triglyceride accumulation was also reduced by JY-2, as determined by Oil Red O staining. In addition, JY-2 restored PA-impaired glucose-stimulated insulin secretion (GSIS), in conjunction with an increased mRNA expression of PDX1, MafA, and insulin in INS-1 cells. The in vivo efficacy of JY-2 was examined using C57BL/6J, db/db, and high fat-diet induced obese and diabetic (DIO) mice models, and showed that JY-2 improved glucose tolerance, in parallel with a reduced mRNA expression of gluconeogenic genes. Pharmacokinetic analysis revealed that JY-2 exhibited excellent oral bioavailability (98%), with little adverse effects. These results demonstrated that the novel FoxO1 inhibitor, JY-2, may exert beneficial anti-diabetic effects and that it warrants further investigation as a novel anti-diabetic drug candidate.
Collapse
Affiliation(s)
- Hye-Eun Choi
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, 21999, South Korea
| | - YuSik Kim
- Severance Institute for Vascular and Metabolic Research Yonsei University School of Medicine, Seoul, 06230, South Korea
| | - Han-Joo Lee
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983, South Korea
| | - Hyae Gyeong Cheon
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, 21999, South Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea.
| |
Collapse
|
14
|
Peng DQ, Smith SB, Lee HG. Vitamin A regulates intramuscular adipose tissue and muscle development: promoting high-quality beef production. J Anim Sci Biotechnol 2021; 12:34. [PMID: 33663602 PMCID: PMC7934359 DOI: 10.1186/s40104-021-00558-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/18/2021] [Indexed: 01/07/2023] Open
Abstract
During growth in cattle, the development of intramuscular adipose tissue and muscle is dependent upon cell hyperplasia (increased number of adipocytes) and hypertrophy (increased size of adipocytes). Based on the results of previous studies, other adipose tissue depots (e.g., perirenal and subcutaneous) develop from the fetal stage primarily as brown adipose tissue. The hyperplastic stage of intramuscular adipose is considered to develop from late pregnancy, but there is no evidence indicating that intramuscular adipose tissue develops initially as brown adipose tissue. Hyperplastic growth of intramuscular adipose continues well into postweaning and is dependent on the timing of the transition to grain-based diets; thereafter, the late-stage development of intramuscular adipose tissue is dominated by hypertrophy. For muscle development, hyperplasia of myoblasts lasts from early (following development of somites in the embryo) to middle pregnancy, after which growth of muscle is the result of hypertrophy of myofibers. Vitamin A is a fat-soluble compound that is required for the normal immunologic function, vision, cellular proliferation, and differentiation. Here we review the roles of vitamin A in intramuscular adipose tissue and muscle development in cattle. Vitamin A regulates both hyperplasia and hypertrophy in in vitro experiments. Vitamin A supplementation at the early stage and restriction at fattening stage generate opposite effects in the beef cattle. Appropriate vitamin A supplementation and restriction strategy increase intramuscular adipose tissue development (i.e., marbling or intramuscular fat) in some in vivo trials. Besides, hyperplasia and hypertrophy of myoblasts/myotubes were affected by vitamin A treatment in in vitro trials. Additionally, some studies reported an interaction between the alcohol dehydrogenase-1C (ADH1C) genotype and vitamin A feed restriction for the development of marbling and/or intramuscular adipose tissue, which was dependent on the timing and level of vitamin A restriction. Therefore, the feed strategy of vitamin A has the visible impact on the marbling and muscle development in the cattle, which will be helpful to promote the quality of the beef.
Collapse
Affiliation(s)
- Dong Qiao Peng
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Stephen B Smith
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Hong Gu Lee
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, South Korea.
| |
Collapse
|
15
|
Henry SA, Crivello S, Nguyen TM, Cybulska M, Hoang NS, Nguyen M, Badial T, Emami N, Awada N, Woodward JF, So CH. G protein-coupled receptor kinase 2 modifies the ability of Caenorhabditis elegans to survive oxidative stress. Cell Stress Chaperones 2021; 26:187-197. [PMID: 33064264 PMCID: PMC7736396 DOI: 10.1007/s12192-020-01168-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/19/2020] [Accepted: 09/22/2020] [Indexed: 01/25/2023] Open
Abstract
Survival and adaptation to oxidative stress is important for many organisms, and these occur through the activation of many different signaling pathways. In this report, we showed that Caenorhabditis (C.) elegans G protein-coupled receptor kinases modified the ability of the organism to resist oxidative stress. In acute oxidative stress studies using juglone, loss-of-function grk-2 mutants were more resistant to oxidative stress compared with loss-of-function grk-1 mutants and the wild-type N2 animals. This effect was Ce-AKT-1 dependent, suggesting that Ce-GRK2 adjusted C. elegans oxidative stress resistance through the IGF/insulin-like signaling (IIS) pathway. Treating C. elegans with a GRK2 inhibitor, the selective serotonin reuptake inhibitor paroxetine, resulted in increased acute oxidative stress resistance compared with another selective serotonin reuptake inhibitor, fluoxetine. In chronic oxidative stress studies with paraquat, both grk-1 and grk-2 mutants had longer lifespan compared with the wild-type N2 animals in stress. In summary, this research showed the importance of both GRKs, especially GRK2, in modifying oxidative stress resistance.
Collapse
Affiliation(s)
- Stacy A Henry
- Roseman University of Health Sciences School of Pharmacy, 11 Sunset Way, Henderson, NV, 89014, USA
| | - Selina Crivello
- Roseman University of Health Sciences School of Pharmacy, 11 Sunset Way, Henderson, NV, 89014, USA
| | - Tina M Nguyen
- Roseman University of Health Sciences School of Pharmacy, 11 Sunset Way, Henderson, NV, 89014, USA
| | - Magdalena Cybulska
- Roseman University of Health Sciences School of Pharmacy, 11 Sunset Way, Henderson, NV, 89014, USA
| | - Ngoc S Hoang
- Roseman University of Health Sciences School of Pharmacy, 11 Sunset Way, Henderson, NV, 89014, USA
| | - Mary Nguyen
- Roseman University of Health Sciences School of Pharmacy, 11 Sunset Way, Henderson, NV, 89014, USA
| | | | - Nazgol Emami
- Roseman University of Health Sciences School of Pharmacy, 11 Sunset Way, Henderson, NV, 89014, USA
| | - Nasma Awada
- Roseman University of Health Sciences School of Pharmacy, 11 Sunset Way, Henderson, NV, 89014, USA
| | - Johnathen F Woodward
- Roseman University of Health Sciences School of Pharmacy, 11 Sunset Way, Henderson, NV, 89014, USA
| | - Christopher H So
- Roseman University of Health Sciences School of Pharmacy, 11 Sunset Way, Henderson, NV, 89014, USA.
| |
Collapse
|
16
|
Webster JM, Kempen LJAP, Hardy RS, Langen RCJ. Inflammation and Skeletal Muscle Wasting During Cachexia. Front Physiol 2020; 11:597675. [PMID: 33329046 PMCID: PMC7710765 DOI: 10.3389/fphys.2020.597675] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022] Open
Abstract
Cachexia is the involuntary loss of muscle and adipose tissue that strongly affects mortality and treatment efficacy in patients with cancer or chronic inflammatory disease. Currently, no specific treatments or interventions are available for patients developing this disorder. Given the well-documented involvement of pro-inflammatory cytokines in muscle and fat metabolism in physiological responses and in the pathophysiology of chronic inflammatory disease and cancer, considerable interest has revolved around their role in mediating cachexia. This has been supported by association studies that report increased levels of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) in some, but not all, cancers and in chronic inflammatory diseases such as chronic obstructive pulmonary disease (COPD) and rheumatoid arthritis (RA). In addition, preclinical studies including animal disease models have provided a substantial body of evidence implicating a causal contribution of systemic inflammation to cachexia. The presence of inflammatory cytokines can affect skeletal muscle through several direct mechanisms, relying on activation of the corresponding receptor expressed by muscle, and resulting in inhibition of muscle protein synthesis (MPS), elevation of catabolic activity through the ubiquitin-proteasomal system (UPS) and autophagy, and impairment of myogenesis. Additionally, systemic inflammatory mediators indirectly contribute to muscle wasting through dysregulation of tissue and organ systems, including GCs via the hypothalamus-pituitary-adrenal (HPA) axis, the digestive system leading to anorexia-cachexia, and alterations in liver and adipocyte behavior, which subsequently impact on muscle. Finally, myokines secreted by skeletal muscle itself in response to inflammation have been implicated as autocrine and endocrine mediators of cachexia, as well as potential modulators of this debilitating condition. While inflammation has been shown to play a pivotal role in cachexia development, further understanding how these cytokines contribute to disease progression is required to reveal biomarkers or diagnostic tools to help identify at risk patients, or enable the design of targeted therapies to prevent or delay the progression of cachexia.
Collapse
Affiliation(s)
- Justine M. Webster
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Laura J. A. P. Kempen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Rowan S. Hardy
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Institute for Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Ramon C. J. Langen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
17
|
Liu W, Li Y, Luo B. Current perspective on the regulation of FOXO4 and its role in disease progression. Cell Mol Life Sci 2020; 77:651-663. [PMID: 31529218 PMCID: PMC11104957 DOI: 10.1007/s00018-019-03297-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/21/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022]
Abstract
Forkhead box O4 (FOXO4) is a member of the FOXO family that regulates a number of genes involved in metabolism, cell cycle, apoptosis, and cellular homeostasis via transcriptional activity. It also mediates cell responses to oxidative stress and treatment with antitumor agents. The expression of FOXO4 is repressed by microRNAs in multiple cancer cells, while FOXO4 function is regulated by post-translational modifications and interaction with other proteins. The deregulation of FOXO4 is closely linked to the progression of several types of cancer, senescence, and other diseases. In this review, we present recent findings on the regulation of FOXO4 in physiological and pathological conditions and provide an overview of the complex role of FOXO4 in disease development and response to therapy.
Collapse
Affiliation(s)
- Wen Liu
- Department of Pathogenic Biology, Faculty of Medicine, Qingdao University, Qingdao, China
| | - Yong Li
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Faculty of Medicine, Qingdao University, Qingdao, China
| | - Bing Luo
- Department of Pathogenic Biology, Faculty of Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
18
|
Exploring the Interface between Inflammatory and Therapeutic Glucocorticoid Induced Bone and Muscle Loss. Int J Mol Sci 2019; 20:ijms20225768. [PMID: 31744114 PMCID: PMC6888251 DOI: 10.3390/ijms20225768] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 02/02/2023] Open
Abstract
Due to their potent immunomodulatory anti-inflammatory properties, synthetic glucocorticoids (GCs) are widely utilized in the treatment of chronic inflammatory disease. In this review, we examine our current understanding of how chronic inflammation and commonly used therapeutic GCs interact to regulate bone and muscle metabolism. Whilst both inflammation and therapeutic GCs directly promote systemic osteoporosis and muscle wasting, the mechanisms whereby they achieve this are distinct. Importantly, their interactions in vivo are greatly complicated secondary to the directly opposing actions of GCs on a wide array of pro-inflammatory signalling pathways that underpin catabolic and anti-anabolic metabolism. Several clinical studies have attempted to address the net effects of therapeutic glucocorticoids on inflammatory bone loss and muscle wasting using a range of approaches. These have yielded a wide array of results further complicated by the nature of inflammatory disease, underlying the disease management and regimen of GC therapy. Here, we report the latest findings related to these pathway interactions and explore the latest insights from murine models of disease aimed at modelling these processes and delineating the contribution of pre-receptor steroid metabolism. Understanding these processes remains paramount in the effective management of patients with chronic inflammatory disease.
Collapse
|
19
|
Pyropia yezoensis Protein Supplementation Prevents Dexamethasone-Induced Muscle Atrophy in C57BL/6 Mice. Mar Drugs 2018; 16:md16090328. [PMID: 30208614 PMCID: PMC6163250 DOI: 10.3390/md16090328] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/05/2018] [Accepted: 09/09/2018] [Indexed: 12/29/2022] Open
Abstract
We investigated the protective effects of Pyropia yezoensis crude protein (PYCP) against dexamethasone (DEX)-induced myotube atrophy and its underlying mechanisms. DEX (3 mg/kg body weight, intraperitoneal injection) and PYCP (150 and 300 mg/kg body weight, oral) were administrated to mice for 18 days, and the effects of PYCP on DEX-induced muscle atrophy were evaluated. Body weight, calf thickness, and gastrocnemius and tibialis anterior muscle weight were significantly decreased by DEX administration (p < 0.05), while PYCP supplementation effectively prevented the DEX-induced decrease in body weight, calf thickness, and muscle weight. PYCP supplementation also attenuated the DEX-induced increase in serum glucose, creatine kinase, and lactate dehydrogenase levels. Additionally, PYCP supplementation reversed DEX-induced muscle atrophy via the regulation of the insulin-like growth factor-I/protein kinase B/rapamycin-sensitive mTOR complex I/forkhead box O signaling pathway. The mechanistic investigation revealed that PYCP inhibited the ubiquitin-proteasome and autophagy-lysosome pathways in DEX-administrated C57BL/6 mice. These findings demonstrated that PYCP increased protein synthesis and decreased protein breakdown to prevent muscle atrophy. Therefore, PYCP supplementation appears to be useful for preventing muscle atrophy.
Collapse
|
20
|
Arcidiacono B, Chiefari E, Messineo S, Bilotta FL, Pastore I, Corigliano DM, Foti DP, Brunetti A. HMGA1 is a novel transcriptional regulator of the FoxO1 gene. Endocrine 2018; 60:56-64. [PMID: 29052178 PMCID: PMC5845622 DOI: 10.1007/s12020-017-1445-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/27/2017] [Indexed: 12/14/2022]
Abstract
PURPOSE The forkhead transcription factor (FoxO1) is a master transcriptional regulator of fundamental cellular processes ranging from cell proliferation and differentiation to inflammation and metabolism. However, despite its relevance, the mechanism(s) underlying FoxO1 gene regulation are largely unknown. We have previously shown that the chromatin factor high-mobility group A1 (HMGA1) plays a key role in the transcriptional regulation of glucose-responsive genes, including some that are involved in FoxO1-mediated glucose metabolism. Here we investigated the impact of HMGA1 on FoxO1 gene expression. METHODS FoxO1 protein and gene expression studies were performed by Western blot analysis combined with qRT-PCR of material from human cultured cells and EBV-transformed lymphoblasts, and from primary cultured hepatocytes from wild-type and Hmga1 -/- mice. Reporter gene assays and chromatin immunoprecipitation for binding of HMGA1 to the endogenous FoxoO1 locus were performed in cells overexpressing HMGA1 and in cells pretreated with siRNA targeting HMGA1. RESULTS HMGA1 increased FoxO1 mRNA and protein expression in vitro, in cultured HepG2 and HEK-293 cells by binding FoxO1 gene promoter, thereby activating FoxO1 gene transcription. Forced expression of HMGA1 in primary cultured hepatocytes from Hmga1 -/- mice and in EBV-transformed lymphoblasts from subjects with reduced expression of endogenous HMGA1 increased FoxO1 mRNA and protein levels. CONCLUSION These findings may contribute to the understanding of FoxO1 gene regulation and its role in metabolism.
Collapse
Affiliation(s)
- Biagio Arcidiacono
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Sebastiano Messineo
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Francesco L Bilotta
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Ida Pastore
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Domenica M Corigliano
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Daniela P Foti
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy.
| |
Collapse
|
21
|
Choi IY, Lee C, Longo VD. Nutrition and fasting mimicking diets in the prevention and treatment of autoimmune diseases and immunosenescence. Mol Cell Endocrinol 2017; 455:4-12. [PMID: 28137612 PMCID: PMC5862044 DOI: 10.1016/j.mce.2017.01.042] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 02/04/2023]
Abstract
Complex and coordinated signals are necessary to initiate and sustain the activation, proliferation, and differentiation of lymphocytes. These signals, which are known to determine T-cell fate and function, also depend on the metabolic state of the organism. Recent studies indicate that both the type and levels of nutrients can influence the generation, survival and function of lymphocytes and therefore can affect several autoimmune diseases. Here, we review the dysregulation of lymphocytes during autoimmunity and aging, the mechanisms associated with loss of immune function, and how fasting mimicking diets and other dietary interventions affect autoimmunity and immunosenescence.
Collapse
Affiliation(s)
- In Young Choi
- Longevity Institute, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; Department of Microbiology, Immunology, Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Changhan Lee
- Longevity Institute, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Valter D Longo
- Longevity Institute, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; Department of Neuroscience, Dana and David Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA 90089, USA; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; IFOM, FIRC Institute of Molecular Oncology, 20139, Milan, Italy.
| |
Collapse
|
22
|
Adaptation to Chronic Nutritional Stress Leads to Reduced Dependence on Microbiota in Drosophila melanogaster. mBio 2017; 8:mBio.01496-17. [PMID: 29066546 PMCID: PMC5654931 DOI: 10.1128/mbio.01496-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Numerous studies have shown that animal nutrition is tightly linked to gut microbiota, especially under nutritional stress. In Drosophila melanogaster, microbiota are known to promote juvenile growth, development, and survival on poor diets, mainly through enhanced digestion leading to changes in hormonal signaling. Here, we show that this reliance on microbiota is greatly reduced in replicated Drosophila populations that became genetically adapted to a poor larval diet in the course of over 170 generations of experimental evolution. Protein and polysaccharide digestion in these poor-diet-adapted populations became much less dependent on colonization with microbiota. This was accompanied by changes in expression levels of dFOXO transcription factor, a key regulator of cell growth and survival, and many of its targets. These evolutionary changes in the expression of dFOXO targets to a large degree mimic the response of the same genes to microbiota, suggesting that the evolutionary adaptation to poor diet acted on mechanisms that normally mediate the response to microbiota. Our study suggests that some metazoans have retained the evolutionary potential to adapt their physiology such that association with microbiota may become optional rather than essential.IMPORTANCE Animals depend on gut microbiota for various metabolic tasks, particularly under conditions of nutritional stress, a relationship usually regarded as an inherent aspect of animal physiology. Here, we use experimental evolution in fly populations to show that the degree of host dependence on microbiota can substantially and rapidly change as the host population evolves in response to poor diet. Our results suggest that, although microbiota may initially greatly facilitate coping with suboptimal diets, chronic nutritional stress experienced over multiple generations leads to evolutionary adaptation in physiology and gut digestive properties that reduces dependence on the microbiota for growth and survival. Thus, despite its ancient evolutionary history, the reliance of animal hosts on their microbial partners can be surprisingly flexible and may be relaxed by short-term evolution.
Collapse
|
23
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic progressive liver disorder that begins with simple hepatic steatosis and progresses to non-alcoholic steatohepatitis, fibrosis, cirrhosis, and even liver cancer. As the global prevalence of NAFLD rises, it is increasingly important that we understand its pathogenesis and develop effective therapies for this chronic disease. Forkhead box O (FOXO) transcription factors are key downstream regulators in the insulin/insulin-like growth factor 1 (IGF1) signaling pathway, and have been implicated in a range of cellular functions including the regulation of glucose, triglyceride, and cholesterol homeostasis. The role of FOXOs in the modulation of immune response and inflammation is complex, with reports of both pro- and anti-inflammatory effects. FOXOs are reported to protect against hepatic fibrosis by inhibiting proliferation and transdifferentiation of hepatic stellate cells. Mice that are deficient in hepatic FOXOs are more susceptible to non-alcoholic steatohepatitis than wild-type controls. In summary, FOXOs play a critical role in maintaining metabolic and cellular homeostasis in the liver, and dysregulation of FOXOs may be involved in NAFLD development.
Collapse
Affiliation(s)
- X Charlie Dong
- Department of Biochemistry and Molecular Biology, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
24
|
Martin-Rincon M, Morales-Alamo D, Calbet JAL. Exercise-mediated modulation of autophagy in skeletal muscle. Scand J Med Sci Sports 2017; 28:772-781. [PMID: 28685860 DOI: 10.1111/sms.12945] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2017] [Indexed: 12/13/2022]
Abstract
Although exercise exerts multiple beneficial health effects, it may also damage cellular structures. Damaged elements are continuously degraded and its constituents recycled to produce renovated structures through a process called autophagy, which is essential for the adaptation to training. Autophagy is particularly active in skeletal muscle, where it can be evaluated using specific molecular markers of activation (unc-51-like kinase 1 [ULK1] phosphorylation) and specific proteins indicating increased autophagosome content (increased total LC3, LC3-II, LC3-II/LC3-I ratio). Studies in humans are technically limited but have provided evidence suggesting the activation of autophagy in skeletal muscle through AMP-activated protein kinase (AMPK) and its downstream target ULK1. Autophagy activation is more likely when the intensity is elevated and the exercise performed in the fasted state. The autophagy-gene program and autophagosome content are upregulated after ultraendurance running competitions. However, autophagosome content is reduced after endurance exercise at moderate intensities (50% and 70% of VO2 max) for 60-120 minutes. Autophagosome content is decreased within the first few hours after resistance training. The effects of regular endurance and strength training on basal autophagy remain to be established in humans. One study has reported that acute severe hypoxia increases autophagosome content in human skeletal muscle, which is reverted by 20 minutes of low-intensity exercise. Experiments with transgenic mice have shown that autophagy is necessary for skeletal muscle adaptation to training. Little is known on how genetic factors, environment, nutrition, drugs and diseases may interact with exercise to modulate autophagy at rest and during exercise in humans.
Collapse
Affiliation(s)
- M Martin-Rincon
- Research Institute of Biomedical and Health Sciences (IUIBS), Las Palmas de Gran Canaria, Canary Islands, Spain.,Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - D Morales-Alamo
- Research Institute of Biomedical and Health Sciences (IUIBS), Las Palmas de Gran Canaria, Canary Islands, Spain.,Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - J A L Calbet
- Research Institute of Biomedical and Health Sciences (IUIBS), Las Palmas de Gran Canaria, Canary Islands, Spain.,Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| |
Collapse
|
25
|
Li Y, Ma Z, Jiang S, Hu W, Li T, Di S, Wang D, Yang Y. A global perspective on FOXO1 in lipid metabolism and lipid-related diseases. Prog Lipid Res 2017; 66:42-49. [PMID: 28392404 DOI: 10.1016/j.plipres.2017.04.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/29/2017] [Accepted: 04/05/2017] [Indexed: 02/07/2023]
Abstract
Lipid metabolism is a complex physiological process that is involved in nutrient adjustment, hormone regulation, and homeostasis. An unhealthy lifestyle and chronic nutrient overload can cause lipid metabolism disorders, which may lead to serious lipid-related diseases, including obesity, non-alcoholic fatty liver disease (NAFLD), and type 2 diabetes mellitus (T2DM). Therefore, tools for preventing dysfunctional lipid metabolism are urgently needed. The transcription factor forkhead box protein O1 (FOXO1) is involved in lipid metabolism and plays a critical role in the development of lipid-related diseases. In this review, we provide a global perspective on the role of FOXO1 in lipid metabolism and lipid-related diseases. The information included here may be useful for the design of future studies and advancing investigations of FOXO1 as a therapeutic target.
Collapse
Affiliation(s)
- Yue Li
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China.
| | - Yang Yang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
26
|
Abstract
Cancer is the second leading cause of death in the USA and among the leading major diseases in the world. It is anticipated to continue to increase because of the growth of the aging population and prevalence of risk factors such as obesity, smoking, and/or poor dietary habits. Cancer treatment has remained relatively similar during the past 30 years with chemotherapy and/or radiotherapy in combination with surgery remaining the standard therapies although novel therapies are slowly replacing or complementing the standard ones. According to the American Cancer Society, the dietary recommendation for cancer patients receiving chemotherapy is to increase calorie and protein intake. In addition, there are no clear guidelines on the type of nutrition that could have a major impact on cancer incidence. Yet, various forms of reduced caloric intake such as calorie restriction (CR) or fasting demonstrate a wide range of beneficial effects able to help prevent malignancies and increase the efficacy of cancer therapies. Whereas chronic CR provides both beneficial and detrimental effects as well as major compliance challenges, periodic fasting (PF), fasting-mimicking diets (FMDs), and dietary restriction (DR) without a reduction in calories are emerging as interventions with the potential to be widely used to prevent and treat cancer. Here, we review preclinical and preliminary clinical studies on dietary restriction and fasting and their role in inducing cellular protection and chemotherapy resistance.
Collapse
|
27
|
Cea LA, Balboa E, Puebla C, Vargas AA, Cisterna BA, Escamilla R, Regueira T, Sáez JC. Dexamethasone-induced muscular atrophy is mediated by functional expression of connexin-based hemichannels. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1891-9. [PMID: 27437607 DOI: 10.1016/j.bbadis.2016.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 06/29/2016] [Accepted: 07/12/2016] [Indexed: 11/29/2022]
Abstract
Long-term treatment with high glucocorticoid doses induces skeletal muscle atrophy. However, the molecular mechanism of such atrophy remains unclear. We evaluated the possible involvement of connexin-based hemichannels (Cx HCs) in muscle atrophy induced by dexamethasone (DEX), a synthetic glucocorticoid, on control (Cx43(fl/fl)Cx45(fl/fl)) and Cx43/Cx45 expression-deficient (Cx43(fl/fl)Cx45(fl/fl):Myo-Cre) skeletal myofibers. Myofibers of Cx43(fl/fl)Cx45(fl/fl) mice treated with DEX (5h) expressed several proteins that form non-selective membrane channels (Cx39, Cx43, Cx45, Panx1, P2X7 receptor and TRPV2). After 5h DEX treatment in vivo, myofibers of Cx43(fl/fl)Cx45(fl/fl) mice showed Evans blue uptake, which was absent in myofibers of Cx43(fl/fl)Cx45(fl/fl):Myo-Cre mice. Similar results were obtained in vitro using ethidium as an HC permeability probe, and DEX-induced dye uptake in control myofibers was blocked by P2X7 receptor inhibitors. DEX also induced a significant increase in basal intracellular Ca(2+) signal and a reduction in resting membrane potential in Cx43(fl/fl)Cx45(fl/fl) myofibers, changes that were not elicited by myofibers deficient in Cx43/Cx45 expression. Moreover, treatment with DEX induced NFκB activation and increased mRNA levels of TNF-α in control but not in Cx43/Cx45 expression-deficient myofibers. Finally, a prolonged DEX treatment (7days) increased atrogin-1 and Murf-1 and reduced the cross sectional area of Cx43(fl/fl)Cx45(fl/fl) myofibers, but these parameters remained unaffected in Cx43(fl/fl)Cx45(fl/fl):Myo-Cre myofibers. Therefore, DEX-induced expression of Cx43 and Cx45 plays a critical role in early sarcolemma changes that lead to atrophy. Consequently, this side effect of chronic glucocorticoid treatment might be avoided by co-administration with a Cx HC blocker.
Collapse
Affiliation(s)
- Luis A Cea
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Elisa Balboa
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos Puebla
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile
| | - Aníbal A Vargas
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile
| | - Bruno A Cisterna
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile
| | - Rosalba Escamilla
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile
| | - Tomás Regueira
- Departamento Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan C Sáez
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
28
|
Abstract
The majority of metastatic breast cancers cannot be cured and present a major public health problem worldwide. Approximately 70% of breast cancers express the estrogen receptor, and endocrine-based therapies have significantly improved patient outcomes. However, the development of endocrine resistance is extremely common. Understanding the molecular pathways that regulate the hormone sensitivity of breast cancer cells is important to improving the efficacy of endocrine therapy. It is becoming clearer that the PI3K-AKT-forkhead box O (FOXO) signaling axis is a key player in the hormone-independent growth of many breast cancers. Constitutive PI3K-AKT pathway activation, a driver of breast cancer growth, causes down-regulation of FOXO tumor suppressor functions. This review will summarize what is currently known about the role of FOXOs in endocrine-resistance mechanisms. It will also suggest potential therapeutic strategies for the restoration of normal FOXO transcriptional activity.
Collapse
Affiliation(s)
- M Bullock
- Hormones and Cancer GroupCancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, Pacific Highway Saint Leonards, Sydney, New South Wales 2065, Australia
| |
Collapse
|
29
|
Liu J, Peng Y, Wang X, Fan Y, Qin C, Shi L, Tang Y, Cao K, Li H, Long J, Liu J. Mitochondrial Dysfunction Launches Dexamethasone-Induced Skeletal Muscle Atrophy via AMPK/FOXO3 Signaling. Mol Pharm 2015; 13:73-84. [DOI: 10.1021/acs.molpharmaceut.5b00516] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jing Liu
- Center
for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical
Information Engineering of Ministry of Education, School of Life Science
and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, China
| | - Yunhua Peng
- Center
for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical
Information Engineering of Ministry of Education, School of Life Science
and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, China
| | - Xun Wang
- Center
for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical
Information Engineering of Ministry of Education, School of Life Science
and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, China
| | - Yingying Fan
- Center
for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical
Information Engineering of Ministry of Education, School of Life Science
and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, China
| | - Chuan Qin
- Center
for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical
Information Engineering of Ministry of Education, School of Life Science
and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, China
| | - Le Shi
- Center
for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical
Information Engineering of Ministry of Education, School of Life Science
and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, China
| | - Ying Tang
- Center
for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical
Information Engineering of Ministry of Education, School of Life Science
and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, China
| | - Ke Cao
- Center
for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical
Information Engineering of Ministry of Education, School of Life Science
and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, China
| | - Hua Li
- Center
for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical
Information Engineering of Ministry of Education, School of Life Science
and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, China
| | - Jiangang Long
- Center
for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical
Information Engineering of Ministry of Education, School of Life Science
and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, China
| | - Jiankang Liu
- Center
for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical
Information Engineering of Ministry of Education, School of Life Science
and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin 300381, China
| |
Collapse
|
30
|
MacKrell JG, Yaden BC, Bullock H, Chen K, Shetler P, Bryant HU, Krishnan V. Molecular targets of androgen signaling that characterize skeletal muscle recovery and regeneration. NUCLEAR RECEPTOR SIGNALING 2015; 13:e005. [PMID: 26457071 PMCID: PMC4599140 DOI: 10.1621/nrs.13005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 09/05/2015] [Indexed: 01/25/2023]
Abstract
The high regenerative capacity of adult skeletal muscle relies on a self-renewing depot of adult stem cells, termed muscle satellite cells (MSCs). Androgens, known mediators of overall body composition and specifically skeletal muscle mass, have been shown to regulate MSCs. The possible overlapping function of androgen regulation of muscle growth and MSC activation has not been carefully investigated with regards to muscle regeneration.Therefore, the aim of this study was to examine coinciding androgen-mediated genetic changes in an in vitro MSC model and clinically relevant in vivo models. A gene signature was established via microarray analysis for androgen-mediated MSC engagement and highlighted several markers including follistatin (FST), IGF-1, C-X-C chemokine receptor 4 (CXCR4), hepatocyte growth factor (HGF) and glucocorticoid receptor (GR). In an in vivo muscle atrophy model, androgen re-supplementation significantly increased muscle size and expression of IGF-1, FST, and HGF, while significantly decreasing expression of GR. Biphasic gene expression profiles over the 7-day re-supplementation period identified temporal androgen regulation of molecular targets involved in satellite cell engagement into myogenesis. In a muscle injury model, removal of androgens resulted in delayed muscle recovery and regeneration. Modifications in the androgen signaling gene signature, along with reduced Pax7 and MyoD expression, suggested that limited MSC activation and increased inflammation contributed to the delayed regeneration. However, enhanced MSC activation in the androgen-deplete mouse injury model was driven by an androgen receptor (AR) agonist. These results provide novel in vitro and in vivo evidence describing molecular targets of androgen signaling, while also increasing support for translational use of AR agonists in skeletal muscle recovery and regeneration.
Collapse
Affiliation(s)
- James G MacKrell
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| | - Benjamin C Yaden
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| | - Heather Bullock
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| | - Keyue Chen
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| | - Pamela Shetler
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| | - Henry U Bryant
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| | - Venkatesh Krishnan
- Musculoskeletal Research (JGM, BCY, HB, PS, HUB, VK), Lead Optimization Biology (KC), Lilly Research Labs, Eli Lilly & Company, Indianapolis, IN, USA
| |
Collapse
|
31
|
Klotz LO, Sánchez-Ramos C, Prieto-Arroyo I, Urbánek P, Steinbrenner H, Monsalve M. Redox regulation of FoxO transcription factors. Redox Biol 2015; 6:51-72. [PMID: 26184557 PMCID: PMC4511623 DOI: 10.1016/j.redox.2015.06.019] [Citation(s) in RCA: 550] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/25/2015] [Accepted: 06/30/2015] [Indexed: 12/19/2022] Open
Abstract
Transcription factors of the forkhead box, class O (FoxO) family are important regulators of the cellular stress response and promote the cellular antioxidant defense. On one hand, FoxOs stimulate the transcription of genes coding for antioxidant proteins located in different subcellular compartments, such as in mitochondria (i.e. superoxide dismutase-2, peroxiredoxins 3 and 5) and peroxisomes (catalase), as well as for antioxidant proteins found extracellularly in plasma (e.g., selenoprotein P and ceruloplasmin). On the other hand, reactive oxygen species (ROS) as well as other stressful stimuli that elicit the formation of ROS, may modulate FoxO activity at multiple levels, including posttranslational modifications of FoxOs (such as phosphorylation and acetylation), interaction with coregulators, alterations in FoxO subcellular localization, protein synthesis and stability. Moreover, transcriptional and posttranscriptional control of the expression of genes coding for FoxOs is sensitive to ROS. Here, we review these aspects of FoxO biology focusing on redox regulation of FoxO signaling, and with emphasis on the interplay between ROS and FoxOs under various physiological and pathophysiological conditions. Of particular interest are the dual role played by FoxOs in cancer development and their key role in whole body nutrient homeostasis, modulating metabolic adaptations and/or disturbances in response to low vs. high nutrient intake. Examples discussed here include calorie restriction and starvation as well as adipogenesis, obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Lars-Oliver Klotz
- Institute of Nutrition, Department of Nutrigenomics, Friedrich-Schiller-Universität Jena, Dornburger Straße 29, 07743 Jena, Germany.
| | - Cristina Sánchez-Ramos
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier, 4, 28029 Madrid, Spain
| | - Ignacio Prieto-Arroyo
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier, 4, 28029 Madrid, Spain
| | - Pavel Urbánek
- Institute of Nutrition, Department of Nutrigenomics, Friedrich-Schiller-Universität Jena, Dornburger Straße 29, 07743 Jena, Germany
| | - Holger Steinbrenner
- Institute of Nutrition, Department of Nutrigenomics, Friedrich-Schiller-Universität Jena, Dornburger Straße 29, 07743 Jena, Germany
| | - Maria Monsalve
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier, 4, 28029 Madrid, Spain.
| |
Collapse
|
32
|
Keogh K, Kenny DA, Kelly AK, Waters SM. Insulin secretion and signaling in response to dietary restriction and subsequent re-alimentation in cattle. Physiol Genomics 2015; 47:344-54. [PMID: 26015430 DOI: 10.1152/physiolgenomics.00002.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 05/22/2015] [Indexed: 01/04/2023] Open
Abstract
The objectives of this study were to examine systemic insulin response to a glucose tolerance test (GTT) and transcript abundance of genes of the insulin signaling pathway in skeletal muscle, during both dietary restriction and re-alimentation-induced compensatory growth. Holstein Friesian bulls were blocked to one of two groups: 1) restricted feed allowance for 125 days (period 1) (RES, n = 15) followed by ad libitum feeding for 55 days (period 2) or 2) ad libitum access to feed throughout (periods 1 and 2) (ADLIB, n = 15). On days 90 and 36 of periods 1 and 2, respectively, a GTT was performed. M. longissimus dorsi biopsies were harvested from all bulls on days 120 and 15 of periods 1 and 2, respectively, and RNA-Seq analysis was performed. RES displayed a lower growth rate during period 1 (RES: 0.6 kg/day, ADLIB: 1.9 kg/day; P < 0.001), subsequently gaining more during re-alimentation (RES: 2.5 kg/day, ADLIB: 1.4 kg/day; P < 0.001). Systemic insulin response to glucose administration was lower in RES in period 1 (P < 0.001) with no difference observed during period 2. The insulin signaling pathway in M. longissimus dorsi was enriched (P < 0.05) in response to dietary restriction but not during re-alimentation (P > 0.05). Genes differentially expressed in the insulin signaling pathway suggested a greater sensitivity to insulin in skeletal muscle, with pleiotropic effects of insulin signaling interrupted during dietary restriction. Collectively, these results indicate increased sensitivity to glucose clearance and skeletal muscle insulin signaling during dietary restriction; however, no overall role for insulin was apparent in expressing compensatory growth.
Collapse
Affiliation(s)
- Kate Keogh
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Dunsany, County Meath, Ireland; and UCD School of Agriculture and Food Science, Belfield, Dublin, Ireland
| | - David A Kenny
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Dunsany, County Meath, Ireland; and
| | - Alan K Kelly
- UCD School of Agriculture and Food Science, Belfield, Dublin, Ireland
| | - Sinéad M Waters
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Dunsany, County Meath, Ireland; and
| |
Collapse
|
33
|
Fang XL, Zhu XT, Chen SF, Zhang ZQ, Zeng QJ, Deng L, Peng JL, Yu JJ, Wang LN, Wang SB, Gao P, Jiang QY, Shu G. Differential gene expression pattern in hypothalamus of chickens during fasting-induced metabolic reprogramming: Functions of glucose and lipid metabolism in the feed intake of chickens. Poult Sci 2014; 93:2841-54. [DOI: 10.3382/ps.2014-04047] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
34
|
Current overview of functions of FoxO proteins, with special regards to cellular homeostasis, cell response to stress, as well as inflammation and aging. Adv Med Sci 2013. [PMID: 23183765 DOI: 10.2478/v10039-012-0039-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
FoxO transcription factors act at the interconnections between metabolic pathways inducible by many important signal transducers and mediators, such as p53, Ikk-β, NFKB, Akt, sirtuins, PTEN, and others. This may account for a crucial significance of disruptions in FoxO functions both in many kinds of diseases (including cancer, chronic inflammatory diseases, degenerative diseases, obesity, polymetabolic syndrome) and in some disease-like conditions (such as inflammaging, cachexia related to chronic inflammation, cancer-promotion by some chronic inflammatory responses, and the aging process itself). This paper reviews complex interactions between FoxOs and other signal transducers, trying to pinpoint how exactly disruptions of FoxO functions may occur, and how they may contribute to occurrence, development or complications of the conditions mentioned above.
Collapse
|
35
|
Glucocorticoid-induced skeletal muscle atrophy. Int J Biochem Cell Biol 2013; 45:2163-72. [PMID: 23806868 DOI: 10.1016/j.biocel.2013.05.036] [Citation(s) in RCA: 438] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 05/27/2013] [Accepted: 05/29/2013] [Indexed: 12/11/2022]
Abstract
Many pathological states characterized by muscle atrophy (e.g., sepsis, cachexia, starvation, metabolic acidosis and severe insulinopenia) are associated with an increase in circulating glucocorticoids (GC) levels, suggesting that GC could trigger the muscle atrophy observed in these conditions. GC-induced muscle atrophy is characterized by fast-twitch, glycolytic muscles atrophy illustrated by decreased fiber cross-sectional area and reduced myofibrillar protein content. GC-induced muscle atrophy results from increased protein breakdown and decreased protein synthesis. Increased muscle proteolysis, in particular through the activation of the ubiquitin proteasome and the lysosomal systems, is considered to play a major role in the catabolic action of GC. The stimulation by GC of these two proteolytic systems is mediated through the increased expression of several Atrogenes ("genes involved in atrophy"), such as FOXO, Atrogin-1, and MuRF-1. The inhibitory effect of GC on muscle protein synthesis is thought to result mainly from the inhibition of the mTOR/S6 kinase 1 pathway. These changes in muscle protein turnover could be explained by changes in the muscle production of two growth factors, namely Insulin-like Growth Factor (IGF)-I, a muscle anabolic growth factor and Myostatin, a muscle catabolic growth factor. This review will discuss the recent progress made in the understanding of the mechanisms involved in GC-induced muscle atrophy and consider the implications of these advancements in the development of new therapeutic approaches for treating GC-induced myopathy. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
|
36
|
Treatment with pharmacological PPARα agonists stimulates the ubiquitin proteasome pathway and myofibrillar protein breakdown in skeletal muscle of rodents. Biochim Biophys Acta Gen Subj 2012; 1830:2105-17. [PMID: 23041501 DOI: 10.1016/j.bbagen.2012.09.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 09/28/2012] [Accepted: 09/28/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND Treatment of hyperlipidemic patients with fibrates, agonists of peroxisome proliferator-activated receptor α (PPARα), provokes muscle atrophy as a side effect. The molecular mechanism underlying this phenomenon is still unknown. We tested the hypothesis that activation of PPARα leads to an up-regulation of the ubiquitin proteasome system (UPS) which plays a major role in protein degradation in muscle. METHODS Rats, wild-type and PPARα-deficient mice (PPARα(-/-)) were treated with synthetic PPARα agonists (clofibrate, WY-14,643) to study their effect on the UPS and myofibrillar protein breakdown in muscle. RESULTS In rats and wild-type mice but not PPARα(-/-) mice, clofibrate or WY-14,643 caused increases in mRNA and protein levels of the ubiquitin ligases atrogin-1 and MuRF1 in muscle. Wild-type mice treated with WY-14,643 had a greater 3-methylhistidine release from incubated muscle and lesser muscle weights. In addition, wild-type mice but not PPARα(-/-) mice treated with WY-14,643 had higher amounts of ubiquitin-protein conjugates, a decreased activity of PI3K/Akt1 signalling, and an increased activity of FoxO1 transcription factor in muscle. Reporter gene and gel shift experiments revealed that the atrogin-1 and MuRF1 promoter do not contain functional PPARα DNA-binding sites. CONCLUSIONS These findings indicate that fibrates stimulate ubiquitination of proteins in skeletal muscle which in turn stimulates protein degradation. Up-regulation of ubiquitin ligases is probably not mediated by PPARα-dependent gene transcription but by PPARα-dependent inhibition of the PI3K/Akt1 signalling pathway leading to activation of FoxO1. GENERAL SIGNIFICANCE PPARα plays a role in the regulation of the ubiquitin proteasome system.
Collapse
|
37
|
McAinch AJ, Lee JS, Bruce CR, Tunstall RJ, Hawley JA, Cameron-Smith D. Dietary Regulation of Fat Oxidative Gene Expression in Different Skeletal Muscle Fiber Types. ACTA ACUST UNITED AC 2012; 11:1471-9. [PMID: 14694211 DOI: 10.1038/oby.2003.197] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE To determine the effect of a high-fat diet on the expression of genes important for fat oxidation, the protein abundance of the transcription factors peroxisome proliferator-activated receptor (PPAR) isoforms alpha and gamma, and selected enzyme activities in type I and II skeletal muscle. RESEARCH METHODS AND PROCEDURES Sprague-Dawley rats consumed either a high-fat (HF: 78% energy, n = 8) or high-carbohydrate (64% energy, n = 8) diet for 8 weeks while remaining sedentary. RESULTS The expression of genes important for fat oxidation tended to increase in both type I (soleus) and type II (extensor digitorum longus) fiber types after an HF dietary intervention. However, the expression of muscle type carnitine palmitoyltransferase I was not increased in extensor digitorum longus. Analysis of the gene expression of both peroxisome proliferator-activated receptor-gamma coactivator and fork-head transcription factor O1 demonstrated no alteration in response to the HF diet. Similarly, PPARalpha and PPARgamma protein levels were also not altered by the HF diet. DISCUSSION An HF diet increased the expression of an array of genes involved in lipid metabolism, with only subtle differences evident in the response within differing skeletal muscle fiber types. Despite changes in gene expression, there were no effects of diet on peroxisome proliferator-activated receptor-gamma coactivator and fork-head transcription factor O1 mRNA and the protein abundance of PPARalpha and PPARgamma.
Collapse
MESH Headings
- 3-Hydroxyacyl CoA Dehydrogenases/genetics
- 3-Hydroxyacyl CoA Dehydrogenases/metabolism
- Animals
- Blotting, Western
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- Carnitine O-Palmitoyltransferase/genetics
- Carnitine O-Palmitoyltransferase/metabolism
- DNA-Binding Proteins
- Dietary Fats/metabolism
- Dietary Fats/pharmacology
- Female
- Forkhead Transcription Factors
- Gene Expression Regulation, Enzymologic/physiology
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiology
- Nerve Tissue Proteins
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Protein Isoforms
- Protein Kinases/genetics
- Protein Kinases/metabolism
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Random Allocation
- Rats
- Rats, Sprague-Dawley
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Andrew J McAinch
- School of Health Sciences, Deakin University, Burwood, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
38
|
Hanaoka BY, Peterson CA, Horbinski C, Crofford LJ. Implications of glucocorticoid therapy in idiopathic inflammatory myopathies. Nat Rev Rheumatol 2012; 8:448-57. [PMID: 22688888 DOI: 10.1038/nrrheum.2012.85] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glucocorticoids are the cornerstone of therapy in patients with idiopathic inflammatory myopathies (IIM), despite adverse effects and suboptimal therapy success rates. Glucocorticoids are used in patients with IIM to suppress inflammatory and immune responses implicated in the pathogenesis of these diseases. Nevertheless, potential inhibitory effects of glucocorticoids on skeletal muscle mass, myogenesis and immune responses that promote skeletal muscle regeneration after muscle injury warrant attention. Glucocorticoids lead to skeletal muscle catabolism by modulating major pathways involved in regulating muscle mass. Glucocorticoids also inhibit muscle regeneration by decreasing myogenic cell proliferation and differentiation. Finally, glucocorticoids might have inhibitory effects on immune cells that have been shown to be an important component of the muscle regenerative response. Better understanding of the signalling pathways involved in restorative versus adverse effects of glucocorticoids in IIM could yield additional insight into the aetiopathogenesis of persistent muscle weakness in patients with IIM after glucocorticoid treatment, and help in the development of novel, targeted treatment options with fewer adverse effects.
Collapse
Affiliation(s)
- Beatriz Y Hanaoka
- Department of Internal Medicine, Division of Rheumatology, University of Kentucky, Room J-509, 740 South Limestone Drive, Lexington, KY 40502, USA
| | | | | | | |
Collapse
|
39
|
Constantin-Teodosiu D, Constantin D, Stephens F, Laithwaite D, Greenhaff PL. The role of FOXO and PPAR transcription factors in diet-mediated inhibition of PDC activation and carbohydrate oxidation during exercise in humans and the role of pharmacological activation of PDC in overriding these changes. Diabetes 2012; 61:1017-24. [PMID: 22315317 PMCID: PMC3331777 DOI: 10.2337/db11-0799] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
High-fat feeding inhibits pyruvate dehydrogenase complex (PDC)-controlled carbohydrate (CHO) oxidation, which contributes to muscle insulin resistance. We aimed to reveal molecular changes underpinning this process in resting and exercising humans. We also tested whether pharmacological activation of PDC overrides these diet-induced changes. Healthy males consumed a control diet (CD) and on two further occasions an isocaloric high-fat diet (HFD). After each diet, subjects cycled for 60 min after intravenous infusion with saline (CD and HFD) or dichloroacetate (HFD+DCA). Quadriceps muscle biopsies obtained before and after 10 and 60 min of exercise were used to estimate CHO use, PDC activation, and mRNAs associated with insulin, fat, and CHO signaling. Compared with CD, HFD increased resting pyruvate dehydrogenase kinase 2 (PDK2), PDK4, forkhead box class O transcription factor 1 (FOXO1), and peroxisome proliferator-activated receptor transcription factor α (PPARα) mRNA and reduced PDC activation. Exercise increased PDC activation and whole-body CHO use in HFD, but to a lower extent than in CD. Meanwhile PDK4 and FOXO1, but not PPARα or PDK2, mRNA remained elevated. HFD+DCA activated PDC throughout and restored whole-body CHO use during exercise. FOXO1 appears to play a role in HFD-mediated muscle PDK4 upregulation and inhibition of PDC and CHO oxidation in humans. Also, pharmacological activation of PDC restores HFD-mediated inhibition of CHO oxidation during exercise.
Collapse
|
40
|
Shi YC, Liao VHC, Pan TM. Monascin from red mold dioscorea as a novel antidiabetic and antioxidative stress agent in rats and Caenorhabditis elegans. Free Radic Biol Med 2012; 52:109-17. [PMID: 22041455 DOI: 10.1016/j.freeradbiomed.2011.09.034] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 09/25/2011] [Accepted: 09/28/2011] [Indexed: 12/19/2022]
Abstract
Monascin is a major yellow compound from red mold dioscorea. We investigated monascin to test whether this compound acts as an antidiabetic and antioxidative stress agent in diabetic rats and Caenorhabditis elegans. The mechanisms by which monascin exerts its action in vivo were also examined. Streptozotocin (STZ)-induced diabetic rats were given monascin at 30 mg/kg/day and sacrificed after 8 weeks. Blood glucose and serum insulin, triglyceride, total cholesterol, and high-density lipoprotein and antioxidative enzymes in the pancreas of rats were measured. In addition, monascin was evaluated for stress resistance and potential associated mechanisms in C. elegans. Throughout the 8-week experimental period, significantly lowered blood glucose, serum triglyceride, and total cholesterol and higher high-density lipoprotein levels were observed in monascin-treated rats. Monascin-treated rats showed higher serum insulin level, lower reactive oxygen species production, and higher activities of glutathione peroxidase, superoxide dismutase, and catalase in the pancreas compared to diabetic control rats. In addition, monascin significantly induced the hepatic mRNA levels of FOXO3a, FOXO1, MnSOD, and catalase in STZ-induced diabetic rats. Monascin-treated C. elegans showed an increased survival rate during oxidative stress and heat stress treatments compared to untreated controls. Moreover, monascin extended the life span under high-glucose conditions and enhanced expression of small heat shock protein (sHSP-16.2), superoxide dismutase (SOD-3), and glutathione S-transferase (GST-4) in C. elegans. Finally, we showed that monascin affected the subcellular distribution of the FOXO transcription factor DAF-16, whereas it was unable to enhance oxidative stress resistance in the daf-16 deletion mutant in C. elegans. Mechanistic studies in rats and C. elegans suggest that the protective effects of monascin are mediated via regulation of the FOXO/DAF-16-dependent insulin signaling pathway by inducing the expression of stress response/antioxidant genes, thereby enhancing oxidative stress resistance.
Collapse
Affiliation(s)
- Yeu-Ching Shi
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | | | | |
Collapse
|
41
|
Zhang N, Guo XF, Wang H, Zhang HS. Determination of amino acids and catecholamines derivatized with 3-(4-chlorobenzoyl)-2-quinolinecarboxaldehyde in PC12 and HEK293 cells by capillary electrophoresis with laser-induced fluorescence detection. Anal Bioanal Chem 2011; 401:297-304. [DOI: 10.1007/s00216-011-5056-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 04/19/2011] [Accepted: 04/22/2011] [Indexed: 11/29/2022]
|
42
|
Lee C, Longo VD. Fasting vs dietary restriction in cellular protection and cancer treatment: from model organisms to patients. Oncogene 2011; 30:3305-16. [PMID: 21516129 DOI: 10.1038/onc.2011.91] [Citation(s) in RCA: 202] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The dietary recommendation for cancer patients receiving chemotherapy, as described by the American Cancer Society, is to increase calorie and protein intake. Yet, in simple organisms, mice, and humans, fasting--no calorie intake--induces a wide range of changes associated with cellular protection, which would be difficult to achieve even with a cocktail of potent drugs. In mammals, the protective effect of fasting is mediated, in part, by an over 50% reduction in glucose and insulin-like growth factor 1 (IGF-I) levels. Because proto-oncogenes function as key negative regulators of the protective changes induced by fasting, cells expressing oncogenes, and therefore the great majority of cancer cells, should not respond to the protective signals generated by fasting, promoting the differential protection (differential stress resistance) of normal and cancer cells. Preliminary reports indicate that fasting for up to 5 days followed by a normal diet, may also protect patients against chemotherapy without causing chronic weight loss. By contrast, the long-term 20 to 40% restriction in calorie intake (dietary restriction, DR), whose effects on cancer progression have been studied extensively for decades, requires weeks-months to be effective, causes much more modest changes in glucose and/or IGF-I levels, and promotes chronic weight loss in both rodents and humans. In this study, we review the basic as well as clinical studies on fasting, cellular protection and chemotherapy resistance, and compare them to those on DR and cancer treatment. Although additional pre-clinical and clinical studies are necessary, fasting has the potential to be translated into effective clinical interventions for the protection of patients and the improvement of therapeutic index.
Collapse
Affiliation(s)
- C Lee
- Andrus Gerontology Center, Department of Biological Sciences and Norris Cancer Center, University of Southern California, Los Angeles, CA 90089-0191, USA
| | | |
Collapse
|
43
|
Abstract
FoxO transcription factors have a conserved role in longevity, and act as tissue-specific tumor suppressors in mammals. Several nodes of interaction have been identified between FoxO transcription factors and p53, a major tumor suppressor in humans and mice. However, the extent and importance of the functional interaction between FoxO and p53 have not been fully explored. Here, we show that p53 regulates the expression of FoxO3, one of the four mammalian FoxO genes, in response to DNA damaging agents in both mouse embryonic fibroblasts and thymocytes. We find that p53 transactivates FoxO3 in cells by binding to a site in the second intron of the FoxO3 gene, a genomic region recently found to be associated with extreme longevity in humans. While FoxO3 is not necessary for p53-dependent cell cycle arrest, FoxO3 appears to modulate p53-dependent apoptosis. We also find that FoxO3 loss does not interact with p53 loss for tumor development in vivo, although the tumor spectrum of p53-deficient mice appears to be affected by FoxO3 loss. Our findings indicate that FoxO3 is a p53 target gene, and suggest that FoxO3 and p53 are part of a regulatory transcriptional network that may have an important role during aging and cancer.
Collapse
|
44
|
Sparks JD, Chamberlain JM, O'Dell C, Khatun I, Hussain MM, Sparks CE. Acute suppression of apo B secretion by insulin occurs independently of MTP. Biochem Biophys Res Commun 2011; 406:252-6. [PMID: 21316344 DOI: 10.1016/j.bbrc.2011.02.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 02/06/2011] [Indexed: 12/12/2022]
Abstract
Secretion of apolipoprotein (apo) B-containing lipoproteins by the liver depends mainly upon apo B availability and microsomal triglyceride transfer protein (MTP) activity and is subject to insulin regulation. Hepatic MTP mRNA expression is negatively regulated by insulin which correlates with inhibition of apo B secretion suggesting that insulin might suppress apo B secretion through an MTP-dependent mechanism. To investigate this possibility, we examined the acute effect of insulin on hepatic MTP expression and activity levels in vivo utilizing apobec-1(-/-) mice. Insulin did not significantly alter hepatic MTP mRNA levels or lipid transfer activity 2h following injection, but suppressed expression of genes important in gluconeogenesis. To study the specific role of MTP, we expressed human MTP (hMTP) in primary rat hepatocytes using adenoviral gene transfer. Increased expression of hMTP resulted in a 47.6±17.9% increase in total apo B secreted. Incubation of hepatocytes with insulin suppressed apo B secretion by 50.1±10.8% in cells over-expressing hMTP and by 53.0±12.4% in control transfected hepatocytes. Results indicate that even under conditions of increased hepatic apo B secretion mediated by MTP, responsiveness of hepatocytes to insulin to suppress apo B secretion is maintained.
Collapse
Affiliation(s)
- Janet D Sparks
- Department of Pathology & Laboratory Medicine, University of Rochester School of Medicine & Dentistry, Box 626, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Protection against dexamethasone-induced muscle atrophy is related to modulation by testosterone of FOXO1 and PGC-1α. Biochem Biophys Res Commun 2010; 403:473-8. [DOI: 10.1016/j.bbrc.2010.11.061] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 11/15/2010] [Indexed: 12/11/2022]
|
46
|
Dentice M, Marsili A, Ambrosio R, Guardiola O, Sibilio A, Paik JH, Minchiotti G, DePinho RA, Fenzi G, Larsen PR, Salvatore D. The FoxO3/type 2 deiodinase pathway is required for normal mouse myogenesis and muscle regeneration. J Clin Invest 2010; 120:4021-30. [PMID: 20978344 PMCID: PMC2964991 DOI: 10.1172/jci43670] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 08/18/2010] [Indexed: 01/14/2023] Open
Abstract
The active thyroid hormone 3,5,3' triiodothyronine (T3) is a major regulator of skeletal muscle function. The deiodinase family of enzymes controls the tissue-specific activation and inactivation of the prohormone thyroxine (T4). Here we show that type 2 deiodinase (D2) is essential for normal mouse myogenesis and muscle regeneration. Indeed, D2-mediated increases in T3 were essential for the enhanced transcription of myogenic differentiation 1 (MyoD) and for execution of the myogenic program. Conversely, the expression of T3-dependent genes was reduced and after injury regeneration markedly delayed in muscles of mice null for the gene encoding D2 (Dio2), despite normal circulating T3 concentrations. Forkhead box O3 (FoxO3) was identified as a key molecule inducing D2 expression and thereby increasing intracellular T3 production. Accordingly, FoxO3-depleted primary myoblasts also had a differentiation deficit that could be rescued by high levels of T3. In conclusion, the FoxO3/D2 pathway selectively enhances intracellular active thyroid hormone concentrations in muscle, providing a striking example of how a circulating hormone can be tissue-specifically activated to influence development locally.
Collapse
Affiliation(s)
- Monica Dentice
- Department of Molecular and Clinical Endocrinology and Oncology, University of Naples “Federico II,” Naples, Italy.
Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
IRCCS Fondazione SDN, Naples, Italy.
Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso,” CNR, Naples, Italy.
Belfer Institute for Applied Cancer Science, Departments of Medical Oncology, Medicine and Genetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
CEINGE–Biotecnologie Avanzate s.c. a r.l., Naples, Italy
| | - Alessandro Marsili
- Department of Molecular and Clinical Endocrinology and Oncology, University of Naples “Federico II,” Naples, Italy.
Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
IRCCS Fondazione SDN, Naples, Italy.
Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso,” CNR, Naples, Italy.
Belfer Institute for Applied Cancer Science, Departments of Medical Oncology, Medicine and Genetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
CEINGE–Biotecnologie Avanzate s.c. a r.l., Naples, Italy
| | - Raffaele Ambrosio
- Department of Molecular and Clinical Endocrinology and Oncology, University of Naples “Federico II,” Naples, Italy.
Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
IRCCS Fondazione SDN, Naples, Italy.
Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso,” CNR, Naples, Italy.
Belfer Institute for Applied Cancer Science, Departments of Medical Oncology, Medicine and Genetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
CEINGE–Biotecnologie Avanzate s.c. a r.l., Naples, Italy
| | - Ombretta Guardiola
- Department of Molecular and Clinical Endocrinology and Oncology, University of Naples “Federico II,” Naples, Italy.
Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
IRCCS Fondazione SDN, Naples, Italy.
Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso,” CNR, Naples, Italy.
Belfer Institute for Applied Cancer Science, Departments of Medical Oncology, Medicine and Genetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
CEINGE–Biotecnologie Avanzate s.c. a r.l., Naples, Italy
| | - Annarita Sibilio
- Department of Molecular and Clinical Endocrinology and Oncology, University of Naples “Federico II,” Naples, Italy.
Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
IRCCS Fondazione SDN, Naples, Italy.
Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso,” CNR, Naples, Italy.
Belfer Institute for Applied Cancer Science, Departments of Medical Oncology, Medicine and Genetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
CEINGE–Biotecnologie Avanzate s.c. a r.l., Naples, Italy
| | - Ji-Hye Paik
- Department of Molecular and Clinical Endocrinology and Oncology, University of Naples “Federico II,” Naples, Italy.
Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
IRCCS Fondazione SDN, Naples, Italy.
Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso,” CNR, Naples, Italy.
Belfer Institute for Applied Cancer Science, Departments of Medical Oncology, Medicine and Genetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
CEINGE–Biotecnologie Avanzate s.c. a r.l., Naples, Italy
| | - Gabriella Minchiotti
- Department of Molecular and Clinical Endocrinology and Oncology, University of Naples “Federico II,” Naples, Italy.
Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
IRCCS Fondazione SDN, Naples, Italy.
Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso,” CNR, Naples, Italy.
Belfer Institute for Applied Cancer Science, Departments of Medical Oncology, Medicine and Genetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
CEINGE–Biotecnologie Avanzate s.c. a r.l., Naples, Italy
| | - Ronald A. DePinho
- Department of Molecular and Clinical Endocrinology and Oncology, University of Naples “Federico II,” Naples, Italy.
Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
IRCCS Fondazione SDN, Naples, Italy.
Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso,” CNR, Naples, Italy.
Belfer Institute for Applied Cancer Science, Departments of Medical Oncology, Medicine and Genetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
CEINGE–Biotecnologie Avanzate s.c. a r.l., Naples, Italy
| | - Gianfranco Fenzi
- Department of Molecular and Clinical Endocrinology and Oncology, University of Naples “Federico II,” Naples, Italy.
Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
IRCCS Fondazione SDN, Naples, Italy.
Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso,” CNR, Naples, Italy.
Belfer Institute for Applied Cancer Science, Departments of Medical Oncology, Medicine and Genetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
CEINGE–Biotecnologie Avanzate s.c. a r.l., Naples, Italy
| | - P. Reed Larsen
- Department of Molecular and Clinical Endocrinology and Oncology, University of Naples “Federico II,” Naples, Italy.
Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
IRCCS Fondazione SDN, Naples, Italy.
Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso,” CNR, Naples, Italy.
Belfer Institute for Applied Cancer Science, Departments of Medical Oncology, Medicine and Genetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
CEINGE–Biotecnologie Avanzate s.c. a r.l., Naples, Italy
| | - Domenico Salvatore
- Department of Molecular and Clinical Endocrinology and Oncology, University of Naples “Federico II,” Naples, Italy.
Thyroid Section, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.
IRCCS Fondazione SDN, Naples, Italy.
Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso,” CNR, Naples, Italy.
Belfer Institute for Applied Cancer Science, Departments of Medical Oncology, Medicine and Genetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
CEINGE–Biotecnologie Avanzate s.c. a r.l., Naples, Italy
| |
Collapse
|
47
|
Buford TW, Anton SD, Judge AR, Marzetti E, Wohlgemuth SE, Carter CS, Leeuwenburgh C, Pahor M, Manini TM. Models of accelerated sarcopenia: critical pieces for solving the puzzle of age-related muscle atrophy. Ageing Res Rev 2010; 9:369-83. [PMID: 20438881 PMCID: PMC3788572 DOI: 10.1016/j.arr.2010.04.004] [Citation(s) in RCA: 203] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 04/14/2010] [Accepted: 04/15/2010] [Indexed: 12/25/2022]
Abstract
Sarcopenia, the age-related loss of skeletal muscle mass, is a significant public health concern that continues to grow in relevance as the population ages. Certain conditions have the strong potential to coincide with sarcopenia to accelerate the progression of muscle atrophy in older adults. Among these conditions are co-morbid diseases common to older individuals such as cancer, kidney disease, diabetes, and peripheral artery disease. Furthermore, behaviors such as poor nutrition and physical inactivity are well-known to contribute to sarcopenia development. However, we argue that these behaviors are not inherent to the development of sarcopenia but rather accelerate its progression. In the present review, we discuss how these factors affect systemic and cellular mechanisms that contribute to skeletal muscle atrophy. In addition, we describe gaps in the literature concerning the role of these factors in accelerating sarcopenia progression. Elucidating biochemical pathways related to accelerated muscle atrophy may allow for improved discovery of therapeutic treatments related to sarcopenia.
Collapse
Affiliation(s)
- Thomas W. Buford
- Institute on Aging, University of Florida, Gainesville, FL 32611
| | - Stephen D. Anton
- Institute on Aging, University of Florida, Gainesville, FL 32611
| | - Andrew R. Judge
- Institute on Aging, University of Florida, Gainesville, FL 32611
| | | | | | | | | | - Marco Pahor
- Institute on Aging, University of Florida, Gainesville, FL 32611
| | - Todd M. Manini
- Institute on Aging, University of Florida, Gainesville, FL 32611
| |
Collapse
|
48
|
Smith GR, Shanley DP. Modelling the response of FOXO transcription factors to multiple post-translational modifications made by ageing-related signalling pathways. PLoS One 2010; 5:e11092. [PMID: 20567500 PMCID: PMC2886341 DOI: 10.1371/journal.pone.0011092] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 05/01/2010] [Indexed: 01/10/2023] Open
Abstract
FOXO transcription factors are an important, conserved family of regulators of cellular processes including metabolism, cell-cycle progression, apoptosis and stress resistance. They are required for the efficacy of several of the genetic interventions that modulate lifespan. FOXO activity is regulated by multiple post-translational modifications (PTMs) that affect its subcellular localization, half-life, DNA binding and transcriptional activity. Here, we show how a mathematical modelling approach can be used to simulate the effects, singly and in combination, of these PTMs. Our model is implemented using the Systems Biology Markup Language (SBML), generated by an ancillary program and simulated in a stochastic framework. The use of the ancillary program to generate the SBML is necessary because the possibility that many regulatory PTMs may be added, each independently of the others, means that a large number of chemically distinct forms of the FOXO molecule must be taken into account, and the program is used to generate them. Although the model does not yet include detailed representations of events upstream and downstream of FOXO, we show how it can qualitatively, and in some cases quantitatively, reproduce the known effects of certain treatments that induce various single and multiple PTMs, and allows for a complex spatiotemporal interplay of effects due to the activation of multiple PTM-inducing treatments. Thus, it provides an important framework to integrate current knowledge about the behaviour of FOXO. The approach should be generally applicable to other proteins experiencing multiple regulations.
Collapse
Affiliation(s)
- Graham R. Smith
- Henry Wellcome Laboratory for Biogerontology, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Daryl P. Shanley
- Henry Wellcome Laboratory for Biogerontology, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, United Kingdom
- * E-mail:
| |
Collapse
|
49
|
Gebhardt R, Hovhannisyan A. Organ patterning in the adult stage: the role of Wnt/beta-catenin signaling in liver zonation and beyond. Dev Dyn 2010; 239:45-55. [PMID: 19705440 DOI: 10.1002/dvdy.22041] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Wnt/beta-catenin signaling has been found to play key roles in metabolic zonation of adult liver, regeneration, and hepatocellular carcinogenesis. In this review, recent progress in this field is summarized, in particular the rapidly growing knowledge about the various interactions of beta-catenin with many transcription factors involved in controlling metabolism. These interactions may provide the basis for understanding how the wide range of activities of Wnt/beta-catenin signaling is differentially interpreted. Based on these results, a three-level mode for the molecular interpretation of beta-catenin activity gradients in liver is proposed favoring cell differentiation, metabolic zonation, and proliferation. While derangement of the combinatorial interplay of the various transcription factors with beta-catenin at the intermediary activity level may contribute to the development of metabolic diseases, extremely high activation of beta-catenin may eventually lead to initiation and progression of hepatocellular tumors.
Collapse
Affiliation(s)
- Rolf Gebhardt
- Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany.
| | | |
Collapse
|
50
|
Cleveland BM, Weber GM. Effects of insulin-like growth factor-I, insulin, and leucine on protein turnover and ubiquitin ligase expression in rainbow trout primary myocytes. Am J Physiol Regul Integr Comp Physiol 2009; 298:R341-50. [PMID: 20007517 DOI: 10.1152/ajpregu.00516.2009] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The effects of insulin-like growth factor-I (IGF-I), insulin, and leucine on protein turnover and pathways that regulate proteolytic gene expression and protein polyubiquitination were investigated in primary cultures of 4-day-old rainbow trout myocytes. Supplementing media with 100 nM IGF-I increased protein synthesis by 13% (P < 0.05) and decreased protein degradation by 14% (P < 0.05). Treatment with 1 microM insulin increased protein synthesis by 13% (P < 0.05) and decreased protein degradation by 17% (P < 0.05). Supplementing media containing 0.6 mM leucine with an additional 2.5 mM leucine did not increase protein synthesis rates but reduced rates of protein degradation by 8% (P < 0.05). IGF-I (1 nM-100 nM) and insulin (1 nM-1 microM) independently reduced the abundance of ubiquitin ligase mRNA in a dose-dependent manner, with maximal reductions of approximately 70% for muscle atrophy F-box (Fbx) 32, 40% for Fbx25, and 25% for muscle RING finger-1 (MuRF1, P < 0.05). IGF-I and insulin stimulated phosphorylation of FOXO1 and FOXO4 (P < 0.05), which was inhibited by the phosphatidylinositol 3-kinase (PI 3-kinase) inhibitor wortmannin, and decreased the abundance of polyubiquitinated proteins by 10-20% (P < 0.05). Supplementing media with leucine reduced Fbx32 expression by 25% (P < 0.05) but did not affect Fbx25 nor MuRF1 transcript abundance. Serum deprivation decreased rates of protein synthesis by 60% (P < 0.05), increased protein degradation by 40% (P < 0.05), and increased expression of all ubiquitin ligases. These data suggest that, similar to mammals, the inhibitory effects of IGF-I and insulin on proteolysis occur via P I3-kinase/protein kinase B signaling and are partially responsible for the ability of these compounds to promote protein accretion.
Collapse
Affiliation(s)
- Beth M Cleveland
- United States Department of Agriculture, Agricultural Research Service, National Center for Cool and Cold Water Aquaculture, Kearneysville, West Virginia, USA.
| | | |
Collapse
|