1
|
Feng Z, Wang Y, Fu Z, Liao J, Liu H, Zhou M. Exploring the Causal Effects of Mineral Metabolism Disorders on Telomere and Mitochondrial DNA: A Bidirectional Two-Sample Mendelian Randomization Analysis. Nutrients 2024; 16:1417. [PMID: 38794655 PMCID: PMC11123946 DOI: 10.3390/nu16101417] [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: 04/11/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
The aim of this study was to assess the causal relationships between mineral metabolism disorders, representative of trace elements, and key aging biomarkers: telomere length (TL) and mitochondrial DNA copy number (mtDNA-CN). Utilizing bidirectional Mendelian randomization (MR) analysis in combination with the two-stage least squares (2SLS) method, we explored the causal relationships between mineral metabolism disorders and these aging indicators. Sensitivity analysis can be used to determine the reliability and robustness of the research results. The results confirmed that a positive causal relationship was observed between mineral metabolism disorders and TL (p < 0.05), while the causal relationship with mtDNA-CN was not significant (p > 0.05). Focusing on subgroup analyses of specific minerals, our findings indicated a distinct positive causal relationship between iron metabolism disorders and both TL and mtDNA-CN (p < 0.05). In contrast, disorders in magnesium and phosphorus metabolism did not exhibit significant causal effects on either aging biomarker (p > 0.05). Moreover, reverse MR analysis did not reveal any significant causal effects of TL and mtDNA-CN on mineral metabolism disorders (p > 0.05). The combination of 2SLS with MR analysis further reinforced the positive causal relationship between iron levels and both TL and mtDNA-CN (p < 0.05). Notably, the sensitivity analysis did not indicate significant pleiotropy or heterogeneity within these causal relationships (p > 0.05). These findings highlight the pivotal role of iron metabolism in cellular aging, particularly in regulating TL and sustaining mtDNA-CN, offering new insights into how mineral metabolism disorders influence aging biomarkers. Our research underscores the importance of trace element balance, especially regarding iron intake, in combating the aging process. This provides a potential strategy for slowing aging through the adjustment of trace element intake, laying the groundwork for future research into the relationship between trace elements and healthy aging.
Collapse
Affiliation(s)
| | | | | | | | | | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China or (Z.F.); (Y.W.); (Z.F.); (J.L.); (H.L.)
| |
Collapse
|
2
|
Hwang MS, Park J, Ham Y, Lee IH, Chun KH. Roles of Protein Post-Translational Modifications During Adipocyte Senescence. Int J Biol Sci 2023; 19:5245-5256. [PMID: 37928271 PMCID: PMC10620833 DOI: 10.7150/ijbs.86404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/27/2023] [Indexed: 11/07/2023] Open
Abstract
Adipocytes are adipose tissues that supply energy to the body through lipids. The two main types of adipocytes comprise white adipocytes (WAT) that store energy, and brown adipocytes (BAT), which generate heat by burning stored fat (thermogenesis). Emerging evidence indicates that dysregulated adipocyte senescence may disrupt metabolic homeostasis, leading to various diseases and aging. Adipocytes undergo senescence via irreversible cell-cycle arrest in response to DNA damage, oxidative stress, telomere dysfunction, or adipocyte over-expansion upon chronic lipid accumulation. The amount of detectable BAT decreases with age. Activation of cell cycle regulators and dysregulation of adipogenesis-regulating factors may constitute a molecular mechanism that accelerates adipocyte senescence. To better understand the regulation of adipocyte senescence, the effects of post-translational modifications (PTMs), is essential for clarifying the activity and stability of these proteins. PTMs are covalent enzymatic protein modifications introduced following protein biosynthesis, such as phosphorylation, acetylation, ubiquitination, or glycosylation. Determining the contribution of PTMs to adipocyte senescence may identify new therapeutic targets for the regulation of adipocyte senescence. In this review, we discuss a conceptual case in which PTMs regulate adipocyte senescence and explain the mechanisms underlying protein regulation, which may lead to the development of effective strategies to combat metabolic diseases.
Collapse
Affiliation(s)
- Min-Seon Hwang
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Institute of Genetic Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jingyeong Park
- Department of Life Science, College of Natural Science, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Yunha Ham
- Department of Life Science, College of Natural Science, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - In Hye Lee
- Department of Life Science, College of Natural Science, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Kyung-Hee Chun
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Institute of Genetic Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
3
|
Yuan Y, Shi Z, Xiong S, Hu R, Song Q, Song Z, Ong SG, Jiang Y. Differential roles of insulin receptor in adipocyte progenitor cells in mice. Mol Cell Endocrinol 2023; 573:111968. [PMID: 37244600 PMCID: PMC10846871 DOI: 10.1016/j.mce.2023.111968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/29/2023]
Abstract
The development of white adipose tissue (WAT) occurs during distinct embryonic and postnatal stages, and it is subsequently maintained throughout life. However, the specific mediators and mechanisms responsible for WAT development during different phases remain unclear. In this study, we investigate the role of the insulin receptor (IR) in regulating adipogenesis and adipocyte function within adipocyte progenitor cells (APCs) during WAT development and homeostasis. We use two in vivo adipose lineage tracking and deletion systems to delete IR either in embryonic APCs or adult APCs, respectively, to explore the specific requirements of IR during WAT development and WAT homeostasis in mice. Our data suggest that IR expression in APCs may not be essential for adult adipocyte differentiation but appears to be crucial for adipose tissue development. We reveal a surprising divergent role of IR in APCs during WAT development and homeostasis.
Collapse
Affiliation(s)
- Yexian Yuan
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Zuoxiao Shi
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, 60612, USA; Department of Pharmaceutical Sciences, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shaolei Xiong
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ruoci Hu
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, 60612, USA; Department of Pharmaceutical Sciences, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Qing Song
- Department of Kinesiology and Nutrition, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, The University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sang-Ging Ong
- Department of Pharmacology and Regenerative Medicine, College of Medicine, The University of Illinois at Chicago, Illinois, 60612, USA; Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, Illinois, 60612, USA
| | - Yuwei Jiang
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, 60612, USA; Department of Pharmaceutical Sciences, The University of Illinois at Chicago, Chicago, IL, 60612, USA; Division of Endocrinology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
4
|
Hedges CP, Shetty B, Broome SC, MacRae C, Koutsifeli P, Buckels EJ, MacIndoe C, Boix J, Tsiloulis T, Matthews BG, Sinha S, Arendse M, Jaiswal JK, Mellor KM, Hickey AJR, Shepherd PR, Merry TL. Dietary supplementation of clinically utilized PI3K p110α inhibitor extends the lifespan of male and female mice. NATURE AGING 2023; 3:162-172. [PMID: 37118113 DOI: 10.1038/s43587-022-00349-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 12/02/2022] [Indexed: 04/30/2023]
Abstract
Diminished insulin and insulin-like growth factor-1 signaling extends the lifespan of invertebrates1-4; however, whether it is a feasible longevity target in mammals is less clear5-12. Clinically utilized therapeutics that target this pathway, such as small-molecule inhibitors of phosphoinositide 3-kinase p110α (PI3Ki), provide a translatable approach to studying the impact of these pathways on aging. Here, we provide evidence that dietary supplementation with the PI3Ki alpelisib from middle age extends the median and maximal lifespan of mice, an effect that was more pronounced in females. While long-term PI3Ki treatment was well tolerated and led to greater strength and balance, negative impacts on common human aging markers, including reductions in bone mass and mild hyperglycemia, were also evident. These results suggest that while pharmacological suppression of insulin receptor (IR)/insulin-like growth factor receptor (IGFR) targets could represent a promising approach to delaying some aspects of aging, caution should be taken in translation to humans.
Collapse
Affiliation(s)
- C P Hedges
- Discipline of Nutrition, School of Medical Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - B Shetty
- Discipline of Nutrition, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - S C Broome
- Discipline of Nutrition, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - C MacRae
- Discipline of Nutrition, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - P Koutsifeli
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - E J Buckels
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - C MacIndoe
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - J Boix
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - T Tsiloulis
- Discipline of Nutrition, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - B G Matthews
- Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - S Sinha
- Department of Pathology, Waikato Hospital, Hamilton, New Zealand
| | - M Arendse
- Department of Pathology, Waikato Hospital, Hamilton, New Zealand
| | - J K Jaiswal
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - K M Mellor
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - A J R Hickey
- Applied Surgery and Metabolism Laboratory, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - P R Shepherd
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - T L Merry
- Discipline of Nutrition, School of Medical Sciences, University of Auckland, Auckland, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
5
|
Mitochondrial function and nutrient sensing pathways in ageing: enhancing longevity through dietary interventions. Biogerontology 2022; 23:657-680. [PMID: 35842501 DOI: 10.1007/s10522-022-09978-7] [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] [Received: 04/07/2022] [Accepted: 06/30/2022] [Indexed: 12/13/2022]
Abstract
Ageing is accompanied by alterations in several biochemical processes, highly influenced by its environment. It is controlled by the interactions at various levels of biological hierarchy. To maintain homeostasis, a number of nutrient sensors respond to the nutritional status of the cell and control its energy metabolism. Mitochondrial physiology is influenced by the energy status of the cell. The alterations in mitochondrial physiology and the network of nutrient sensors result in mitochondrial damage leading to age related metabolic degeneration and diseases. Calorie restriction (CR) has proved to be as the most successful intervention to achieve the goal of longevity and healthspan. CR elicits a hormetic response and regulates metabolism by modulating these networks. In this review, the authors summarize the interdependent relationship between mitochondrial physiology and nutrient sensors during the ageing process and their role in regulating metabolism.
Collapse
|
6
|
Abdellatif M, Trummer-Herbst V, Heberle AM, Humnig A, Pendl T, Durand S, Cerrato G, Hofer SJ, Islam M, Voglhuber J, Ramos Pittol JM, Kepp O, Hoefler G, Schmidt A, Rainer PP, Scherr D, von Lewinski D, Bisping E, McMullen JR, Diwan A, Eisenberg T, Madeo F, Thedieck K, Kroemer G, Sedej S. Fine-Tuning Cardiac Insulin-Like Growth Factor 1 Receptor Signaling to Promote Health and Longevity. Circulation 2022; 145:1853-1866. [PMID: 35616058 PMCID: PMC9203038 DOI: 10.1161/circulationaha.122.059863] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/20/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND The insulin-like growth factor 1 (IGF1) pathway is a key regulator of cellular metabolism and aging. Although its inhibition promotes longevity across species, the effect of attenuated IGF1 signaling on cardiac aging remains controversial. METHODS We performed a lifelong study to assess cardiac health and lifespan in 2 cardiomyocyte-specific transgenic mouse models with enhanced versus reduced IGF1 receptor (IGF1R) signaling. Male mice with human IGF1R overexpression or dominant negative phosphoinositide 3-kinase mutation were examined at different life stages by echocardiography, invasive hemodynamics, and treadmill coupled to indirect calorimetry. In vitro assays included cardiac histology, mitochondrial respiration, ATP synthesis, autophagic flux, and targeted metabolome profiling, and immunoblots of key IGF1R downstream targets in mouse and human explanted failing and nonfailing hearts, as well. RESULTS Young mice with increased IGF1R signaling exhibited superior cardiac function that progressively declined with aging in an accelerated fashion compared with wild-type animals, resulting in heart failure and a reduced lifespan. In contrast, mice with low cardiac IGF1R signaling exhibited inferior cardiac function early in life, but superior cardiac performance during aging, and increased maximum lifespan, as well. Mechanistically, the late-life detrimental effects of IGF1R activation correlated with suppressed autophagic flux and impaired oxidative phosphorylation in the heart. Low IGF1R activity consistently improved myocardial bioenergetics and function of the aging heart in an autophagy-dependent manner. In humans, failing hearts, but not those with compensated hypertrophy, displayed exaggerated IGF1R expression and signaling activity. CONCLUSIONS Our findings indicate that the relationship between IGF1R signaling and cardiac health is not linear, but rather biphasic. Hence, pharmacological inhibitors of the IGF1 pathway, albeit unsuitable for young individuals, might be worth considering in older adults.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- Department of Cardiology (M.A., V.T.-H., A.H., J.V., A.S., P.P.R., D.S., D.v.L. E.B., S.S.), Medical University of Graz, Austria
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France (M.A., S.D., G.C., O.K., G.K.)
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France (M.A., S.D., G.C., O.K., G.K.)
- BioTechMed Graz, Austria (M.A., S.J.H., J.V., G.H., P.P.R., T.E., F.M., S.S.)
| | - Viktoria Trummer-Herbst
- Department of Cardiology (M.A., V.T.-H., A.H., J.V., A.S., P.P.R., D.S., D.v.L. E.B., S.S.), Medical University of Graz, Austria
| | - Alexander Martin Heberle
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Austria (A.M.H., J.M.R.P., K.T.)
| | - Alina Humnig
- Department of Cardiology (M.A., V.T.-H., A.H., J.V., A.S., P.P.R., D.S., D.v.L. E.B., S.S.), Medical University of Graz, Austria
| | - Tobias Pendl
- Institute of Molecular Biosciences, NAWI Graz (T.P., S.J.H., T.E., F.M.), Washington University School of Medicine, Saint Louis, MO
| | - Sylvère Durand
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France (M.A., S.D., G.C., O.K., G.K.)
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France (M.A., S.D., G.C., O.K., G.K.)
| | - Giulia Cerrato
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France (M.A., S.D., G.C., O.K., G.K.)
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France (M.A., S.D., G.C., O.K., G.K.)
| | - Sebastian J. Hofer
- BioTechMed Graz, Austria (M.A., S.J.H., J.V., G.H., P.P.R., T.E., F.M., S.S.)
- Institute of Molecular Biosciences, NAWI Graz (T.P., S.J.H., T.E., F.M.), Washington University School of Medicine, Saint Louis, MO
- Field of Excellence BioHealth (S.J.H., T.E., F.M.), Washington University School of Medicine, Saint Louis, MO
| | - Moydul Islam
- University of Graz, Austria. Department of Chemistry (M.I.), Washington University School of Medicine, Saint Louis, MO
- Center for Cardiovascular Research and Cardiovascular Division, Department of Medicine (M.I., A.D.), Washington University School of Medicine, Saint Louis, MO
| | - Julia Voglhuber
- BioTechMed Graz, Austria (M.A., S.J.H., J.V., G.H., P.P.R., T.E., F.M., S.S.)
| | - José Miguel Ramos Pittol
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Austria (A.M.H., J.M.R.P., K.T.)
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France (M.A., S.D., G.C., O.K., G.K.)
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France (M.A., S.D., G.C., O.K., G.K.)
| | - Gerald Hoefler
- Diagnostic and Research Center for Molecular BioMedicine, Diagnostic and Research Institute of Pathology (G.H.), Medical University of Graz, Austria
- BioTechMed Graz, Austria (M.A., S.J.H., J.V., G.H., P.P.R., T.E., F.M., S.S.)
| | - Albrecht Schmidt
- Department of Cardiology (M.A., V.T.-H., A.H., J.V., A.S., P.P.R., D.S., D.v.L. E.B., S.S.), Medical University of Graz, Austria
| | - Peter P. Rainer
- Department of Cardiology (M.A., V.T.-H., A.H., J.V., A.S., P.P.R., D.S., D.v.L. E.B., S.S.), Medical University of Graz, Austria
- BioTechMed Graz, Austria (M.A., S.J.H., J.V., G.H., P.P.R., T.E., F.M., S.S.)
| | - Daniel Scherr
- Department of Cardiology (M.A., V.T.-H., A.H., J.V., A.S., P.P.R., D.S., D.v.L. E.B., S.S.), Medical University of Graz, Austria
| | - Dirk von Lewinski
- Department of Cardiology (M.A., V.T.-H., A.H., J.V., A.S., P.P.R., D.S., D.v.L. E.B., S.S.), Medical University of Graz, Austria
| | - Egbert Bisping
- Department of Cardiology (M.A., V.T.-H., A.H., J.V., A.S., P.P.R., D.S., D.v.L. E.B., S.S.), Medical University of Graz, Austria
| | - Julie R. McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.R.M.)
| | - Abhinav Diwan
- Center for Cardiovascular Research and Cardiovascular Division, Department of Medicine (M.I., A.D.), Washington University School of Medicine, Saint Louis, MO
- John Cochran Veterans Affairs Medical Center, Saint Louis, MO (A.D.)
| | - Tobias Eisenberg
- BioTechMed Graz, Austria (M.A., S.J.H., J.V., G.H., P.P.R., T.E., F.M., S.S.)
- Institute of Molecular Biosciences, NAWI Graz (T.P., S.J.H., T.E., F.M.), Washington University School of Medicine, Saint Louis, MO
- Field of Excellence BioHealth (S.J.H., T.E., F.M.), Washington University School of Medicine, Saint Louis, MO
| | - Frank Madeo
- BioTechMed Graz, Austria (M.A., S.J.H., J.V., G.H., P.P.R., T.E., F.M., S.S.)
- Institute of Molecular Biosciences, NAWI Graz (T.P., S.J.H., T.E., F.M.), Washington University School of Medicine, Saint Louis, MO
- Field of Excellence BioHealth (S.J.H., T.E., F.M.), Washington University School of Medicine, Saint Louis, MO
| | - Kathrin Thedieck
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Austria (A.M.H., J.M.R.P., K.T.)
- Department of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, The Netherlands (K.T.)
- Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany (K.T.)
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France (M.A., S.D., G.C., O.K., G.K.)
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France (M.A., S.D., G.C., O.K., G.K.)
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, France (G.K.)
| | - Simon Sedej
- Department of Cardiology (M.A., V.T.-H., A.H., J.V., A.S., P.P.R., D.S., D.v.L. E.B., S.S.), Medical University of Graz, Austria
- BioTechMed Graz, Austria (M.A., S.J.H., J.V., G.H., P.P.R., T.E., F.M., S.S.)
- Institute of Physiology, Faculty of Medicine, University of Maribor, Slovenia (S.S.)
| |
Collapse
|
7
|
Amorim JA, Coppotelli G, Rolo AP, Palmeira CM, Ross JM, Sinclair DA. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat Rev Endocrinol 2022; 18:243-258. [PMID: 35145250 PMCID: PMC9059418 DOI: 10.1038/s41574-021-00626-7] [Citation(s) in RCA: 373] [Impact Index Per Article: 124.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/11/2022]
Abstract
Organismal ageing is accompanied by progressive loss of cellular function and systemic deterioration of multiple tissues, leading to impaired function and increased vulnerability to death. Mitochondria have become recognized not merely as being energy suppliers but also as having an essential role in the development of diseases associated with ageing, such as neurodegenerative and cardiovascular diseases. A growing body of evidence suggests that ageing and age-related diseases are tightly related to an energy supply and demand imbalance, which might be alleviated by a variety of interventions, including physical activity and calorie restriction, as well as naturally occurring molecules targeting conserved longevity pathways. Here, we review key historical advances and progress from the past few years in our understanding of the role of mitochondria in ageing and age-related metabolic diseases. We also highlight emerging scientific innovations using mitochondria-targeted therapeutic approaches.
Collapse
Affiliation(s)
- João A Amorim
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
- Center for Neurosciences and Cell Biology of the University of Coimbra, Coimbra, Portugal
- IIIUC, Institute of Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Giuseppe Coppotelli
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
- George and Anne Ryan Institute for Neuroscience, College of Pharmacy, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Anabela P Rolo
- Center for Neurosciences and Cell Biology of the University of Coimbra, Coimbra, Portugal
- Department of Life Sciences of the University of Coimbra, Coimbra, Portugal
| | - Carlos M Palmeira
- Center for Neurosciences and Cell Biology of the University of Coimbra, Coimbra, Portugal
- Department of Life Sciences of the University of Coimbra, Coimbra, Portugal
| | - Jaime M Ross
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
- George and Anne Ryan Institute for Neuroscience, College of Pharmacy, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - David A Sinclair
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Lin J, Yang Q, Guo J, Li M, Hao Z, He J, Li J. Gut Microbiome Alterations and Hepatic Metabolic Flexibility in the Gansu Zokor, Eospalax cansus: Adaptation to Hypoxic Niches. Front Cardiovasc Med 2022; 9:814076. [PMID: 35402538 PMCID: PMC8984292 DOI: 10.3389/fcvm.2022.814076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
The Gansu zokor (Eospalax cansus), a typical subterranean rodent endemic to the Chinese Loess Plateau, spends almost its whole life in its self-constructed underground burrows and has strong adaptability to ambient hypoxia. Energy adaptation is the key to supporting hypoxia tolerance, and recent studies have shown that the intestinal microbiota has an evident effect on energy metabolism. However, how the gut microbiome of Gansu zokor will change in response to hypoxia and the metabolic role played by the microbiome have not been reported. Thus, we exposed Gansu zokors to severe hypoxia of 6.5% of O2 (6 or 44 h) or moderate hypoxia of 10.5% of O2 (44 h or 4 weeks), and then analyzed 16S rRNA sequencing, metagenomic sequencing, metagenomic binning, liver carbohydrate metabolites, and the related molecular levels. Our results showed that the hypoxia altered the microbiota composition of Gansu zokor, and the relative contribution of Ileibacterium to carbohydrate metabolism became increased under hypoxia, such as glycolysis and fructose metabolism. Furthermore, Gansu zokor liver enhanced carbohydrate metabolism under the short-term (6 or 44 h) hypoxia but it was suppressed under the long-term (4 weeks) hypoxia. Interestingly, under all hypoxia conditions, Gansu zokor liver exhibited enhanced fructose-driven metabolism through increased expression of the GLUT5 fructose transporter, ketohexokinase (KHK), aldolase B (ALDOB), and aldolase C (ALDOC), as well as increased KHK enzymatic activity and fructose utilization. Overall, our results suggest that the altered gut microbiota mediates the carbohydrate metabolic pattern under hypoxia, possibly contributing to the hepatic metabolic flexibility in Gansu zokor, which leads to better adaptation to hypoxic environments.
Collapse
|
9
|
Mishra A, Mirzaei H, Guidi N, Vinciguerra M, Mouton A, Linardic M, Rappa F, Barone R, Navarrete G, Wei M, Brandhorst S, Di Biase S, Morgan TE, Ram Kumar S, Conti PS, Pellegrini M, Bernier M, de Cabo R, Longo VD. Fasting-mimicking diet prevents high-fat diet effect on cardiometabolic risk and lifespan. Nat Metab 2021; 3:1342-1356. [PMID: 34650272 DOI: 10.1038/s42255-021-00469-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 09/02/2021] [Indexed: 12/13/2022]
Abstract
Diet-induced obesity is a major risk factor for metabolic syndrome, diabetes and cardiovascular disease. Here, we show that a 5-d fasting-mimicking diet (FMD), administered every 4 weeks for a period of 2 years, ameliorates the detrimental changes caused by consumption of a high-fat, high-calorie diet (HFCD) in female mice. We demonstrate that monthly FMD cycles inhibit HFCD-mediated obesity by reducing the accumulation of visceral and subcutaneous fat without causing loss of lean body mass. FMD cycles increase cardiac vascularity and function and resistance to cardiotoxins, prevent HFCD-dependent hyperglycaemia, hypercholesterolaemia and hyperleptinaemia and ameliorate impaired glucose and insulin tolerance. The effect of monthly FMD cycles on gene expression associated with mitochondrial metabolism and biogenesis in adipocytes and the sustained ketogenesis in HFCD-fed mice indicate a role for fat cell reprogramming in obesity prevention. These effects of an FMD on adiposity and cardiac ageing could explain the protection from HFCD-dependent early mortality.
Collapse
Affiliation(s)
- Amrendra Mishra
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Hamed Mirzaei
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Novella Guidi
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Manlio Vinciguerra
- International Clinical Research Center, St Anne's University Hospital, Brno, Czech Republic
| | - Alice Mouton
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA, USA
| | - Marina Linardic
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Francesca Rappa
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Rosario Barone
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Gerardo Navarrete
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Min Wei
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Sebastian Brandhorst
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Stefano Di Biase
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Todd E Morgan
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - S Ram Kumar
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Peter S Conti
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Michel Bernier
- Translational Gerontology Branch, Intramural Research Program of the National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, Intramural Research Program of the National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Valter D Longo
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
- IFOM, FIRC Institute of Molecular Oncology, Milano, Italy.
| |
Collapse
|
10
|
Satoh T. Bird evolution by insulin resistance. Trends Endocrinol Metab 2021; 32:803-813. [PMID: 34446347 DOI: 10.1016/j.tem.2021.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/28/2021] [Accepted: 07/26/2021] [Indexed: 10/20/2022]
Abstract
Drift of oxygen concentrations in the atmosphere was one of the main drivers of the evolution of vertebrates. The drop in oxygen concentrations at the Permian-Triassic (PT) boundary may have been the biggest challenge to vertebrates. This hypoxic condition forced theropods to lose certain genes to maximize their efficiency of oxygen usage. Recent studies show that omentin and insulin-sensitive glucose transporter 4 (GLUT4) are missing in the bird genome. Since these gene products play essential roles in maintaining insulin sensitivity, this loss forced theropods to become insulin resistant. Insulin resistance may have been the key to allowing theropods to become hyperathletic under hypoxic conditions and to outcompete mammals during the Triassic period. A second challenge was the gradual increase in oxygen concentrations during the late Jurassic, Cretaceous, and Tertiary periods when reactive oxygen species (ROS) leakage from mitochondria became a problem. Since the simplest solution was the expansion of body size, some theropods became bigger to reduce ROS leakage per volume. Another solution was the development of a constitutively active countermeasure against ROS. A recent study shows that Neoaves have constitutively active nuclear factor erythroid 2-related factor 2 (NRF2) due to deletion of the C-terminal part of the KEAP1 protein, thus allowing Neoaves to express antioxidant enzymes to overcome ROS leakage.
Collapse
Affiliation(s)
- Takumi Satoh
- Department of Anti-Aging Food Research, School of Bioscience and Biotechnology, Tokyo University of Technology, 1404-1 Katakura, Hachioji 192-0982, Japan.
| |
Collapse
|
11
|
Guo F, Seldin M, Péterfy M, Charugundla S, Zhou Z, Lee SD, Mouton A, Rajbhandari P, Zhang W, Pellegrini M, Tontonoz P, Lusis AJ, Shih DM. NOTUM promotes thermogenic capacity and protects against diet-induced obesity in male mice. Sci Rep 2021; 11:16409. [PMID: 34385484 PMCID: PMC8361163 DOI: 10.1038/s41598-021-95720-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/28/2021] [Indexed: 11/29/2022] Open
Abstract
We recently showed that NOTUM, a liver-secreted Wnt inhibitor, can acutely promote browning of white adipose. We now report studies of chronic overexpression of NOTUM in liver indicating that it protects against diet-induced obesity and improves glucose homeostasis in mice. Adeno-associated virus (AAV) vectors were used to overexpress GFP or mouse Notum in the livers of male C57BL/6J mice and the mice were fed an obesifying diet. After 14 weeks of high fat, high sucrose diet feeding, the AAV-Notum mice exhibited decreased obesity and improved glucose tolerance compared to the AAV-GFP mice. Gene expression and immunoblotting analysis of the inguinal fat and brown fat revealed increased expression of beige/brown adipocyte markers in the AAV-Notum group, suggesting enhanced thermogenic capacity by NOTUM. A β3 adrenergic receptor agonist-stimulated lipolysis test suggested increased lipolysis capacity by NOTUM. The levels of collagen and C–C motif chemokine ligand 2 (CCL2) in the epididymal white adipose tissue of the AAV-Notum mice were significantly reduced, suggesting decreased fibrosis and inflammation, respectively. RNA sequencing analysis of inguinal white adipose of 4-week chow diet-fed mice revealed a highly significant enrichment of extracellular matrix (ECM) functional cluster among the down-regulated genes in the AAV-Notum group, suggesting a potential mechanism contributing to improved glucose homeostasis. Our in vitro studies demonstrated that recombinant human NOTUM protein blocked the inhibitory effects of WNT3A on brown adipocyte differentiation. Furthermore, NOTUM attenuated WNT3A’s effects on upregulation of TGF-β signaling and its downstream targets. Overall, our data suggest that NOTUM modulates adipose tissue function by promoting thermogenic capacity and inhibiting fibrosis through inhibition of Wnt signaling.
Collapse
Affiliation(s)
- Fangfei Guo
- Department of Microbiology, Immunology, and Molecular Genetics, Division of Cardiology, Department of Medicine, Department of Human Genetics, University of California, 10833 Le Conte Avenue, A2-237 CHS, Los Angeles, CA, 90095-1679, USA
| | - Marcus Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, CA, 92697, USA
| | - Miklós Péterfy
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Sarada Charugundla
- Department of Microbiology, Immunology, and Molecular Genetics, Division of Cardiology, Department of Medicine, Department of Human Genetics, University of California, 10833 Le Conte Avenue, A2-237 CHS, Los Angeles, CA, 90095-1679, USA
| | - Zhiqiang Zhou
- Department of Microbiology, Immunology, and Molecular Genetics, Division of Cardiology, Department of Medicine, Department of Human Genetics, University of California, 10833 Le Conte Avenue, A2-237 CHS, Los Angeles, CA, 90095-1679, USA
| | - Stephen D Lee
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Alice Mouton
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA, 90095, USA
| | - Prashant Rajbhandari
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine Mount Sinai, New York, NY, 10029, USA
| | - Wenchao Zhang
- Department of Microbiology, Immunology, and Molecular Genetics, Division of Cardiology, Department of Medicine, Department of Human Genetics, University of California, 10833 Le Conte Avenue, A2-237 CHS, Los Angeles, CA, 90095-1679, USA.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Department of Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Matteo Pellegrini
- Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Aldons J Lusis
- Department of Microbiology, Immunology, and Molecular Genetics, Division of Cardiology, Department of Medicine, Department of Human Genetics, University of California, 10833 Le Conte Avenue, A2-237 CHS, Los Angeles, CA, 90095-1679, USA
| | - Diana M Shih
- Department of Microbiology, Immunology, and Molecular Genetics, Division of Cardiology, Department of Medicine, Department of Human Genetics, University of California, 10833 Le Conte Avenue, A2-237 CHS, Los Angeles, CA, 90095-1679, USA.
| |
Collapse
|
12
|
Kirstein AS, Kehr S, Nebe M, Hanschkow M, Barth LAG, Lorenz J, Penke M, Breitfeld J, Le Duc D, Landgraf K, Körner A, Kovacs P, Stadler PF, Kiess W, Garten A. PTEN regulates adipose progenitor cell growth, differentiation, and replicative aging. J Biol Chem 2021; 297:100968. [PMID: 34273354 PMCID: PMC8350019 DOI: 10.1016/j.jbc.2021.100968] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/17/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor suppressor phosphatase and tensin homolog (PTEN) negatively regulates the insulin signaling pathway. Germline PTEN pathogenic variants cause PTEN hamartoma tumor syndrome (PHTS), associated with lipoma development in children. Adipose progenitor cells (APCs) lose their capacity to differentiate into adipocytes during continuous culture, whereas APCs from lipomas of patients with PHTS retain their adipogenic potential over a prolonged period. It remains unclear which mechanisms trigger this aberrant adipose tissue growth. To investigate the role of PTEN in adipose tissue development, we performed functional assays and RNA-Seq of control and PTEN knockdown APCs. Reduction of PTEN levels using siRNA or CRISPR led to enhanced proliferation and differentiation of APCs. Forkhead box protein O1 (FOXO1) transcriptional activity is known to be regulated by insulin signaling, and FOXO1 was downregulated at the mRNA level while its inactivation through phosphorylation increased. FOXO1 phosphorylation initiates the expression of the lipogenesis-activating transcription factor sterol regulatory element-binding protein 1 (SREBP1). SREBP1 levels were higher after PTEN knockdown and may account for the observed enhanced adipogenesis. To validate this, we overexpressed constitutively active FOXO1 in PTEN CRISPR cells and found reduced adipogenesis, accompanied by SREBP1 downregulation. We observed that PTEN CRISPR cells showed less senescence compared with controls and the senescence marker CDKN1A (p21) was downregulated in PTEN knockdown cells. Cellular senescence was the most significantly enriched pathway found in RNA-Seq of PTEN knockdown versus control cells. These results provide evidence that PTEN is involved in the regulation of APC proliferation, differentiation, and senescence, thereby contributing to aberrant adipose tissue growth in patients with PHTS.
Collapse
Affiliation(s)
- Anna S Kirstein
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany.
| | - Stephanie Kehr
- Bioinformatics Group, Department of Computer Science and Interdisciplinary Center for Bioinformatics, Leipzig University, Leipzig, Germany
| | - Michèle Nebe
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Martha Hanschkow
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Lisa A G Barth
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Judith Lorenz
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Melanie Penke
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Jana Breitfeld
- Medical Department III-Endocrinology, Nephrology, Rheumatology, Leipzig University Medical Center, Leipzig, Germany
| | - Diana Le Duc
- Institute of Human Genetics, Leipzig University Medical Center, Leipzig, Germany; Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Kathrin Landgraf
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Antje Körner
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Peter Kovacs
- Medical Department III-Endocrinology, Nephrology, Rheumatology, Leipzig University Medical Center, Leipzig, Germany
| | - Peter F Stadler
- Bioinformatics Group, Department of Computer Science and Interdisciplinary Center for Bioinformatics, Leipzig University, Leipzig, Germany; Max Planck Institute for Mathematics in the Sciences, Leipzig, Germany
| | - Wieland Kiess
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany
| | - Antje Garten
- University Hospital for Children & Adolescents, Center for Pediatric Research, Leipzig University, Leipzig, Germany; Institute for Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
13
|
Abstract
CONTEXT Obesity and type 2 diabetes are associated with chronic hyperinsulinemia, elevated plasma levels of dipeptidyl peptidase-4 (DPP4), and a pro-atherosclerotic milieu. EVIDENCE ACQUISITION PubMed search of the term "insulin and atherosclerosis," "hyperinsulinemia," "atherosclerosis," or "cardiovascular outcomes" cross-referenced with "DPP4." Relevant research and review articles were reviewed. EVIDENCE SYNTHESIS Hyperinsulinemia in the setting of insulin resistance promotes vascular inflammation, vascular smooth muscle cell growth, pathological cholesterol profile, hypertension, and recruitment of immune cells to the endothelium, all contributing to atherosclerosis. DPP4 has pleiotropic functions and its activity is elevated in obese humans. DPP4 mirrors hyperinsulinemia's atherogenic actions in the insulin resistant state, and genetic deletion of DPP4 protects rodents from developing insulin resistance and improves cardiovascular outcomes. DPP4 inhibition in pro-atherosclerotic preclinical models results in reduced inflammation and oxidative stress, improved endothelial function, and decreased atherosclerosis. Increased incretin levels may have contributed to but do not completely account for these benefits. Small clinical studies with DPP4 inhibitors demonstrate reduced carotid intimal thickening, improved endothelial function, and reduced arterial stiffness. To date, this has not been translated to cardiovascular risk reduction for individuals with type 2 diabetes with prior or exaggerated risk of cardiovascular disease. CONCLUSION DPP4 may represent a key link between central obesity, insulin resistance, and atherosclerosis. The gaps in knowledge in DPP4 function and discrepancy in cardiovascular outcomes observed in preclinical and large-scale randomized controlled studies with DPP4 inhibitors warrant additional research.
Collapse
Affiliation(s)
- Kaitlin M Love
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA, USA
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA, USA
| |
Collapse
|
14
|
Zhang AM, Wellberg EA, Kopp JL, Johnson JD. Hyperinsulinemia in Obesity, Inflammation, and Cancer. Diabetes Metab J 2021; 45:285-311. [PMID: 33775061 PMCID: PMC8164941 DOI: 10.4093/dmj.2020.0250] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
The relative insufficiency of insulin secretion and/or insulin action causes diabetes. However, obesity and type 2 diabetes mellitus can be associated with an absolute increase in circulating insulin, a state known as hyperinsulinemia. Studies are beginning to elucidate the cause-effect relationships between hyperinsulinemia and numerous consequences of metabolic dysfunctions. Here, we review recent evidence demonstrating that hyperinsulinemia may play a role in inflammation, aging and development of cancers. In this review, we will focus on the consequences and mechanisms of excess insulin production and action, placing recent findings that have challenged dogma in the context of the existing body of literature. Where relevant, we elaborate on the role of specific signal transduction components in the actions of insulin and consequences of chronic hyperinsulinemia. By discussing the involvement of hyperinsulinemia in various metabolic and other chronic diseases, we may identify more effective therapeutics or lifestyle interventions for preventing or treating obesity, diabetes and cancer. We also seek to identify pertinent questions that are ripe for future investigation.
Collapse
Affiliation(s)
- Anni M.Y. Zhang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth A. Wellberg
- Department of Pathology, University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Harold Hamm Diabetes Center, Oklahoma City, OK, USA
| | - Janel L. Kopp
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - James D. Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
15
|
Mutlu AS, Duffy J, Wang MC. Lipid metabolism and lipid signals in aging and longevity. Dev Cell 2021; 56:1394-1407. [PMID: 33891896 DOI: 10.1016/j.devcel.2021.03.034] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/05/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Lipids play crucial roles in regulating aging and longevity. In the past few decades, a series of genetic pathways have been discovered to regulate lifespan in model organisms. Interestingly, many of these regulatory pathways are linked to lipid metabolism and lipid signaling. Lipid metabolic enzymes undergo significant changes during aging and are regulated by different longevity pathways. Lipids also actively modulate lifespan and health span as signaling molecules. In this review, we summarize recent insights into the roles of lipid metabolism and lipid signaling in aging and discuss lipid-related interventions in promoting longevity.
Collapse
Affiliation(s)
- Ayse Sena Mutlu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jonathon Duffy
- Developmental Biology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meng C Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Developmental Biology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
16
|
Prolonged treatment with a PI3K p110α inhibitor causes sex- and tissue-dependent changes in antioxidant content, but does not affect mitochondrial function. Biosci Rep 2021; 40:226564. [PMID: 33006363 PMCID: PMC7569204 DOI: 10.1042/bsr20201128] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 12/31/2022] Open
Abstract
Genetic inhibition of the p110α isoform of phosphatidylinositol-3-kinase (PI3K) can increase murine lifespan, enhance mitochondrial function and alter tissue-specific oxidative balance. Here, we investigated whether pharmacological inhibition of the p110α isoform of PI3K induces similar enhancement of mitochondrial function in middle-aged mice. Eight-month-old male and female mice were fed a diet containing 0.3 g/kg of the p110α-selective inhibitor BYL-719 (BYL) or a vehicle diet (VEH) for 6 weeks. Mice consuming BYL-719 had higher blood glucose and insulin, and tended towards decreased body weight. After 72 h, gene expression of the mitochondrial biogenesis mediators Pgc1α, Tfam and Nrf1 was greater in liver of BYL-719 males only, but unchanged in skeletal muscle of either sex. Six weeks of BYL-719 treatment did not affect mitochondrial content or function in the liver or skeletal muscle of either sex. In livers of males only, the expression of the antioxidant genes Nfe2l2, Cat, Sod1 and Sod2 increased within 72 h of BYL-719 treatment, and remained higher after 6 weeks. This was associated with an increase in hepatic GSH content and catalase protein expression, and lower H2O2 levels. Our results suggest that pharmacological inhibition of p110α in adult mice does not affect liver or skeletal muscle mitochondrial function, but does show sex- and tissue-specific effects on up-regulation of antioxidant response.
Collapse
|
17
|
Chung KW. Advances in Understanding of the Role of Lipid Metabolism in Aging. Cells 2021; 10:cells10040880. [PMID: 33924316 PMCID: PMC8068994 DOI: 10.3390/cells10040880] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
During aging, body adiposity increases with changes in the metabolism of lipids and their metabolite levels. Considering lipid metabolism, excess adiposity with increased lipotoxicity leads to various age-related diseases, including cardiovascular disease, cancer, arthritis, type 2 diabetes, and Alzheimer's disease. However, the multifaceted nature and complexities of lipid metabolism make it difficult to delineate its exact mechanism and role during aging. With advances in genetic engineering techniques, recent studies have demonstrated that changes in lipid metabolism are associated with aging and age-related diseases. Lipid accumulation and impaired fatty acid utilization in organs are associated with pathophysiological phenotypes of aging. Changes in adipokine levels contribute to aging by modulating changes in systemic metabolism and inflammation. Advances in lipidomic techniques have identified changes in lipid profiles that are associated with aging. Although it remains unclear how lipid metabolism is regulated during aging, or how lipid metabolites impact aging, evidence suggests a dynamic role for lipid metabolism and its metabolites as active participants of signaling pathways and regulators of gene expression. This review describes recent advances in our understanding of lipid metabolism in aging, including established findings and recent approaches.
Collapse
Affiliation(s)
- Ki Wung Chung
- College of Pharmacy, Pusan National University, Busan 46214, Korea
| |
Collapse
|
18
|
Abstract
Aging is characterized by a progressive loss of physiological function leading to increase in the vulnerability to death. This deterioration process occurs in all living organisms and is the primary risk factor for pathological conditions including obesity, type 2 diabetes mellitus, Alzheimer's disease and cardiovascular diseases. Most of the age-related diseases have been associated with impairment of action of an important hormone, namely insulin. It is well-known that this hormone is a critical mediator of metabolism, growth, proliferation and differentiation. Insulin action depends on two processes that determine its circulating levels, insulin secretion and clearance, and insulin sensitivity in its target tissues. Aging has deleterious effects on these three mechanisms, impairing insulin action, thereby increasing the risk for diseases and death. Thus, improving insulin action may be an important strategy to have a healthier and longer life.
Collapse
|
19
|
Adipose PTEN acts as a downstream mediator of a brain-fat axis in environmental enrichment. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2020; 4. [PMID: 35355831 PMCID: PMC8963210 DOI: 10.1016/j.cpnec.2020.100013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background/Objectives Environmental enrichment (EE) is a physiological model to investigate brain-fat interactions. We previously discovered that EE activates the hypothalamic-sympathoneural adipocyte (HSA) axis via induction of brain-derived neurotrophic factor (BDNF), thus leading to sympathetic stimulation of white adipose tissue (WAT) and an anti-obesity phenotype. Here, we investigate whether PTEN acts as a downstream mediator of the HSA axis in the EE. Methods Mice were housed in EE for 4- and 16-week periods to determine how EE regulates adipose PTEN. Hypothalamic injections of adeno-associated viral (AAV) vectors expressing BDNF and a dominant negative form of its receptor were performed to assess the role of the HSA axis in adipose PTEN upregulation. A β-blocker, propranolol, and a denervation agent, 6-hydroydopamine, were administered to assess sympathetic signaling in the observed EE-PTEN phenotype. To determine whether inducing PTEN is sufficient to reproduce certain EE adipose remodeling, we overexpressed PTEN in WAT using an AAV vector. To determine whether adipose PTEN is necessary for the EE-mediated reduction in adipocyte size, we injected a rAAV vector expressing Cre recombinase to the WAT of adult PTENflox mice and placed the mice in EE. Results EE upregulated adipose PTEN expression, which was associated with suppression of AKT and ERK phosphorylation, increased hormone-sensitive lipase (HSL) phosphorylation, and reduced adiposity. PTEN regulation was found to be controlled by the HSA axis—with the hypothalamic BDNF acting as the upstream mediator—and dependent on sympathetic innervation. AAV-mediated adipose PTEN overexpression recapitulated EE-mediated adipose changes including suppression of AKT and ERK phosphorylation, increased HSL phosphorylation, and reduced adipose mass, whereas PTEN knockdown blocked the EE-induced reduction of adipocyte size. Conclusions These data suggest that adipose PTEN responds to environmental stimuli and serves as downstream mediator of WAT remodeling in the EE paradigm, resulting in decreased adipose mass and decreased adipocyte size. Environmental enrichment (EE) induces adipose PTEN expression and is associated with (1) suppression of AKT phosphorylation, (2) increased hormone-sensitive lipase phosphorylation, and (3) decreased adiposity The hypothalamic-sympathoneural-adipocyte (HSA) axis mediates EE-induced adipose PTEN rAAV-mediated gene delivery of PTEN to adipose tissues mimics EE-related adipose remodeling Knockdown of adipose PTEN blocks EE-induced reductions in adipocyte size
Collapse
|
20
|
Botezelli JD, Overby P, Lindo L, Wang S, Haïda O, Lim GE, Templeman NM, Pauli JR, Johnson JD. Adipose depot-specific upregulation of Ucp1 or mitochondrial oxidative complex proteins are early consequences of genetic insulin reduction in mice. Am J Physiol Endocrinol Metab 2020; 319:E529-E539. [PMID: 32715748 DOI: 10.1152/ajpendo.00128.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Hyperinsulinemia plays a causal role in adipose tissue expansion. Mice with reduced insulin have increased energy expenditure, but the mechanisms remained unclear. Here we investigated the effects of genetically reducing insulin production on uncoupling and oxidative mitochondrial proteins in liver, skeletal muscle, white adipose tissue (WAT), and brown adipose tissue (BAT). Male Ins1+/+ or Ins1+/- littermates were fed either a low-fat diet (LFD) or a high-fat diet (HFD) for 4 wk, starting at 8 wk of age. Replicating our previous observations, HFD increased fasting hyperinsulinemia, and Ins1+/- mice had significantly lower circulating insulin compared with Ins1+/+ littermates. Fasting glucose and body weight were not different between genotypes. We did not observe robust significant differences in liver or skeletal muscle. In mesenteric WAT, Ins1+/- mice had reduced Ndufb8 and Sdhb, while Ucp1 was increased in the context of HFD. HFD alone had a dramatic inhibitory effect on Pparg abundance. In inguinal WAT, Ins1+/- mice exhibited significant increases in oxidative complex proteins, independent of diet, without affecting Ucp1, Pparg, or Prdm16:Pparg association. In BAT, lowered insulin increased Sdhb protein levels that had been reduced by HFD. Ucp1 protein, Prdm16:Pparg association, and Sirt3 abundance were all increased in the absence of diet-induced hyperinsulinemia. Our data show that reducing insulin upregulates oxidative proteins in inguinal WAT without affecting Ucp1, whereas in mesenteric WAT and BAT, reducing insulin upregulates Ucp1 in the context of HFD. Preventing hyperinsulinemia has early depot-specific effects on adipose tissue metabolism and helps explain the increased energy expenditure previously reported in Ins1+/- mice.
Collapse
Affiliation(s)
- Jose Diego Botezelli
- Department of Cellular Physiological Sciences, Diabetes Research group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Laboratory of Molecular Biology of Exercise (LaBMEx), Faculty of Applied Sciences, University of Campinas, Limeira, São Paulo, Brazil
| | - Peter Overby
- Department of Cellular Physiological Sciences, Diabetes Research group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lorenzo Lindo
- Department of Cellular Physiological Sciences, Diabetes Research group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Su Wang
- Department of Cellular Physiological Sciences, Diabetes Research group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Obélia Haïda
- Department of Cellular Physiological Sciences, Diabetes Research group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gareth E Lim
- Cardiometabolic axis, Centre de recherche du Centre hospitalier de l'Université de Montréal, Université of Montréal, Montréal, Quebec, Canada
| | | | - Jose Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), Faculty of Applied Sciences, University of Campinas, Limeira, São Paulo, Brazil
| | - James D Johnson
- Department of Cellular Physiological Sciences, Diabetes Research group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
21
|
Hassan QN, Queen NJ, Cao L. Regulation of aging and cancer by enhanced environmental activation of a hypothalamic-sympathoneural-adipocyte axis. Transl Cancer Res 2020; 9:5687-5699. [PMID: 33134111 PMCID: PMC7595574 DOI: 10.21037/tcr.2020.02.39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/11/2020] [Indexed: 12/20/2022]
Abstract
Social and environmental factors impact cancer and energy balance profoundly. Years ago, our lab established the existence of a novel brain-fat interaction we termed the "hypothalamic-sympathoneural-adipocyte (HSA) axis", through which complex environmental stimuli provided by an enriched environment regulate body composition, energy balance, and development of cancer. We have spent a significant portion of the past decade to further characterize the broad health benefits of an enriched environment (for example, leanness, enhanced immune function, and cancer resistance), and to identify mediators in the brain and periphery along the HSA axis. This review summarizes our recent work regarding the interface between endocrinology, immunology, cancer biology, aging, and neuroscience. We will discuss the interplay between these systemic phenomena and how the HSA axis can be targeted for regulation of cancer and aging.
Collapse
Affiliation(s)
- Quais N. Hassan
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Medical Scientist Training Program, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Nicholas J. Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
22
|
Mori MA. Aging: a New Perspective on an Old Issue. AN ACAD BRAS CIENC 2020; 92:e20200437. [PMID: 32638871 DOI: 10.1590/0001-3765202020200437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/21/2020] [Indexed: 12/23/2022] Open
Abstract
The world is undergoing a profound demographic change with a rapid increase in the prevalence of aged individuals. The finitude of life, the burden of senescence and the search for strategies to prolong human life span have troubled humanity since ancient times. However, only in the past few decades we started to understand how organisms age and how life span can be manipulated. Here I give an historical perspective of the aging field and conclude with the notion that aging is controlled by signals from the adipose tissue which are tightly controlled by small non-coding RNAs such as miRNAs.
Collapse
Affiliation(s)
- Marcelo A Mori
- Laboratory of Aging Biology (LaBE), Universidade Estadual de Campinas/UNICAMP, Campinas, SP, Brazil
| |
Collapse
|
23
|
Gonzalez-Gil AM, Elizondo-Montemayor L. The Role of Exercise in the Interplay between Myokines, Hepatokines, Osteokines, Adipokines, and Modulation of Inflammation for Energy Substrate Redistribution and Fat Mass Loss: A Review. Nutrients 2020; 12:E1899. [PMID: 32604889 PMCID: PMC7353393 DOI: 10.3390/nu12061899] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/18/2020] [Accepted: 06/18/2020] [Indexed: 12/17/2022] Open
Abstract
Exercise is an effective strategy for preventing and treating obesity and its related cardiometabolic disorders, resulting in significant loss of body fat mass, white adipose tissue browning, redistribution of energy substrates, optimization of global energy expenditure, enhancement of hypothalamic circuits that control appetite-satiety and energy expenditure, and decreased systemic inflammation and insulin resistance. Novel exercise-inducible soluble factors, including myokines, hepatokines, and osteokines, and immune cytokines and adipokines are hypothesized to play an important role in the body's response to exercise. To our knowledge, no review has provided a comprehensive integrative overview of these novel molecular players and the mechanisms involved in the redistribution of metabolic fuel during and after exercise, the loss of weight and fat mass, and reduced inflammation. In this review, we explain the potential role of these exercise-inducible factors, namely myokines, such as irisin, IL-6, IL-15, METRNL, BAIBA, and myostatin, and hepatokines, in particular selenoprotein P, fetuin A, FGF21, ANGPTL4, and follistatin. We also describe the function of osteokines, specifically osteocalcin, and of adipokines such as leptin, adiponectin, and resistin. We also emphasize an integrative overview of the pleiotropic mechanisms, the metabolic pathways, and the inter-organ crosstalk involved in energy expenditure, fat mass loss, reduced inflammation, and healthy weight induced by exercise.
Collapse
Affiliation(s)
- Adrian M. Gonzalez-Gil
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey N.L. 64710, Mexico;
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Ave. Morones Prieto 300, Monterrey N.L. 64710, Mexico
| | - Leticia Elizondo-Montemayor
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey N.L. 64710, Mexico;
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Ave. Morones Prieto 300, Monterrey N.L. 64710, Mexico
- Tecnologico de Monterrey, Cardiovascular and Metabolomics Research Group, Hospital Zambrano Hellion, San Pedro Garza Garcia P.C. 66278, Mexico
| |
Collapse
|
24
|
Queen NJ, Hassan QN, Cao L. Improvements to Healthspan Through Environmental Enrichment and Lifestyle Interventions: Where Are We Now? Front Neurosci 2020; 14:605. [PMID: 32655354 PMCID: PMC7325954 DOI: 10.3389/fnins.2020.00605] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Environmental enrichment (EE) is an experimental paradigm that is used to explore how a complex, stimulating environment can impact overall health. In laboratory animal experiments, EE housing conditions typically include larger-than-standard cages, abundant bedding, running wheels, mazes, toys, and shelters which are rearranged regularly to further increase stimulation. EE has been shown to improve multiple aspects of health, including but not limited to metabolism, learning and cognition, anxiety and depression, and immunocompetence. Recent advances in lifespan have led some researchers to consider aging as a risk factor for disease. As such, there is a pressing need to understand the processes by which healthspan can be increased. The natural and predictable changes during aging can be reversed or decreased through EE and its underlying mechanisms. Here, we review the use of EE in laboratory animals to understand mechanisms involved in aging, and comment on relative areas of strength and weakness in the current literature. We additionally address current efforts toward applying EE-like lifestyle interventions to human health to extend healthspan. Although increasing lifespan is a clear goal of medical research, improving the quality of this added time also deserves significant attention. Despite hurdles in translating experimental results toward clinical application, we argue there is great potential in using features of EE toward improving human healthy life expectancy or healthspan, especially in the context of increased global longevity.
Collapse
Affiliation(s)
- Nicholas J. Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Quais N. Hassan
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
- Medical Scientist Training Program, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
25
|
Bettedi L, Yan A, Schuster E, Alic N, Foukas LC. Increased mitochondrial and lipid metabolism is a conserved effect of Insulin/PI3K pathway downregulation in adipose tissue. Sci Rep 2020; 10:3418. [PMID: 32099025 PMCID: PMC7042323 DOI: 10.1038/s41598-020-60210-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 01/31/2020] [Indexed: 11/28/2022] Open
Abstract
The Insulin/IGF-1 signalling (IIS) pathway plays an essential role in the regulation of glucose and lipid homeostasis. At the same time, a reduction in the IIS pathway activity can extend lifespan and healthspan in various model organisms. Amongst a number of body organs that sense and respond to insulin/IGF-1, the adipose tissue has a central role in both the metabolic and lifespan effects of IIS at the organismal level. Genetic inactivation of IIS components specifically in the adipose tissue has been shown before to improve metabolic profile and extend lifespan in various model organisms. We sought to identify conserved molecular mechanisms that may underlie the beneficial effects of IIS inhibition in the adipose tissue, specifically at the level of phosphoinositide 3-kinase (PI3K), a key IIS effector molecule. To this end, we inactivated PI3K by genetic means in the fly fat body and by pharmacological inhibition in mammalian adipocytes. Gene expression studies revealed changes to metabolism and upregulation of mitochondrial activity in mouse adipocytes and fly fat bodies with downregulated PI3K, which were confirmed by biochemical assays in mammalian adipocytes. These data suggest that PI3K inactivation has a conserved effect of upregulating mitochondrial metabolism in both fly and mammalian adipose tissue, which likely contributes to the health- and life-span extending effect of IIS pathway downregulation.
Collapse
Affiliation(s)
- Lucia Bettedi
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK.,National Institutes of Child Health and Human Development (NICHD), Bethesda, MD, 20814, USA
| | - Anqi Yan
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK
| | - Eugene Schuster
- Endocrinology Team, Breast Cancer Now, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Nazif Alic
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK
| | - Lazaros C Foukas
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
26
|
Johnson AA, Stolzing A. The role of lipid metabolism in aging, lifespan regulation, and age-related disease. Aging Cell 2019; 18:e13048. [PMID: 31560163 PMCID: PMC6826135 DOI: 10.1111/acel.13048] [Citation(s) in RCA: 258] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/11/2019] [Accepted: 09/04/2019] [Indexed: 12/18/2022] Open
Abstract
An emerging body of data suggests that lipid metabolism has an important role to play in the aging process. Indeed, a plethora of dietary, pharmacological, genetic, and surgical lipid‐related interventions extend lifespan in nematodes, fruit flies, mice, and rats. For example, the impairment of genes involved in ceramide and sphingolipid synthesis extends lifespan in both worms and flies. The overexpression of fatty acid amide hydrolase or lysosomal lipase prolongs life in Caenorhabditis elegans, while the overexpression of diacylglycerol lipase enhances longevity in both C. elegans and Drosophila melanogaster. The surgical removal of adipose tissue extends lifespan in rats, and increased expression of apolipoprotein D enhances survival in both flies and mice. Mouse lifespan can be additionally extended by the genetic deletion of diacylglycerol acyltransferase 1, treatment with the steroid 17‐α‐estradiol, or a ketogenic diet. Moreover, deletion of the phospholipase A2 receptor improves various healthspan parameters in a progeria mouse model. Genome‐wide association studies have found several lipid‐related variants to be associated with human aging. For example, the epsilon 2 and epsilon 4 alleles of apolipoprotein E are associated with extreme longevity and late‐onset neurodegenerative disease, respectively. In humans, blood triglyceride levels tend to increase, while blood lysophosphatidylcholine levels tend to decrease with age. Specific sphingolipid and phospholipid blood profiles have also been shown to change with age and are associated with exceptional human longevity. These data suggest that lipid‐related interventions may improve human healthspan and that blood lipids likely represent a rich source of human aging biomarkers.
Collapse
|
27
|
Mitochondria in the signaling pathways that control longevity and health span. Ageing Res Rev 2019; 54:100940. [PMID: 31415807 PMCID: PMC7479635 DOI: 10.1016/j.arr.2019.100940] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/09/2019] [Accepted: 08/06/2019] [Indexed: 12/26/2022]
Abstract
Genetic and pharmacological intervention studies have identified evolutionarily conserved and functionally interconnected networks of cellular energy homeostasis, nutrient-sensing, and genome damage response signaling pathways, as prominent regulators of longevity and health span in various species. Mitochondria are the primary sites of ATP production and are key players in several other important cellular processes. Mitochondrial dysfunction diminishes tissue and organ functional performance and is a commonly considered feature of the aging process. Here we review the evidence that through reciprocal and multilevel functional interactions, mitochondria are implicated in the lifespan modulation function of these pathways, which altogether constitute a highly dynamic and complex system that controls the aging process. An important characteristic of these pathways is their extensive crosstalk and apparent malleability to modification by non-invasive pharmacological, dietary, and lifestyle interventions, with promising effects on lifespan and health span in animal models and potentially also in humans.
Collapse
|
28
|
Abstract
Reduction of insulin/insulin-like growth factor 1 (IGF1) signaling (IIS) extends the lifespan of various species. So far, several longevity mouse models have been developed containing mutations related to growth signaling deficiency by targeting growth hormone (GH), IGF1, IGF1 receptor, insulin receptor, and insulin receptor substrate. In addition, p70 ribosomal protein S6 kinase 1 (S6K1) knockout leads to lifespan extension. S6K1 encodes an important kinase in the regulation of cell growth. S6K1 is regulated by mechanistic target of rapamycin (mTOR) complex 1. The v-myc myelocytomatosis viral oncogene homolog (MYC)-deficient mice also exhibits a longevity phenotype. The gene expression profiles of these mice models have been measured to identify their longevity mechanisms. Here, we summarize our knowledge of long-lived mouse models related to growth and discuss phenotypic characteristics, including organ-specific gene expression patterns.
Collapse
Affiliation(s)
- Seung-Soo Kim
- Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841, Korea
| | - Cheol-Koo Lee
- Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841; Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02481, Korea
| |
Collapse
|
29
|
Araiz C, Yan A, Bettedi L, Samuelson I, Virtue S, McGavigan AK, Dani C, Vidal-Puig A, Foukas LC. Enhanced β-adrenergic signalling underlies an age-dependent beneficial metabolic effect of PI3K p110α inactivation in adipose tissue. Nat Commun 2019; 10:1546. [PMID: 30948720 PMCID: PMC6449391 DOI: 10.1038/s41467-019-09514-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/12/2019] [Indexed: 01/28/2023] Open
Abstract
The insulin/IGF-1 signalling pathway is a key regulator of metabolism and the rate of ageing. We previously documented that systemic inactivation of phosphoinositide 3-kinase (PI3K) p110α, the principal PI3K isoform that positively regulates insulin signalling, results in a beneficial metabolic effect in aged mice. Here we demonstrate that deletion of p110α specifically in the adipose tissue leads to less fat accumulation over a significant part of adult life and allows the maintenance of normal glucose tolerance despite insulin resistance. This effect of p110α inactivation is due to a potentiating effect on β-adrenergic signalling, which leads to increased catecholamine-induced energy expenditure in the adipose tissue. Our findings provide a paradigm of how partial inactivation of an essential component of the insulin signalling pathway can have an overall beneficial metabolic effect and suggest that PI3K inhibition could potentiate the effect of β-adrenergic agonists in the treatment of obesity and its associated comorbidities.
Collapse
Affiliation(s)
- Caroline Araiz
- Institute of Healthy Ageing & Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK
| | - Anqi Yan
- Institute of Healthy Ageing & Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK
| | - Lucia Bettedi
- Institute of Healthy Ageing & Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK
- National Institutes of Child Health and Human Development (NICHD), Bethesda, MD, 20814, USA
| | - Isabella Samuelson
- Institute of Healthy Ageing & Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Sam Virtue
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Anne K McGavigan
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Christian Dani
- Université Côte d'Azur, CNRS, Inserm, iBV, Faculté de Médecine, 06107, Nice Cedex 2, France
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
- Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK
| | - Lazaros C Foukas
- Institute of Healthy Ageing & Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
30
|
Folch J, Busquets O, Ettcheto M, Sánchez-López E, Pallàs M, Beas-Zarate C, Marin M, Casadesus G, Olloquequi J, Auladell C, Camins A. Experimental Models for Aging and their Potential for Novel Drug Discovery. Curr Neuropharmacol 2018; 16:1466-1483. [PMID: 28685671 PMCID: PMC6295931 DOI: 10.2174/1570159x15666170707155345] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/22/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023] Open
Abstract
Background: An interesting area of scientific research is the development of potential antiaging drugs. In order to pursue this goal, it is necessary to gather the specific knowledge about the adequate preclinical models that are available to evaluate the beneficial effects of new potential drugs. This review is focused on invertebrate and vertebrate preclinical models used to evaluate the efficacy of antiaging compounds, with the objective to extend life span and health span. Methods: Research and online content related to aging, antiaging drugs, experimental aging models is reviewed. Moreover, in this review, the main experimental preclinical models of organisms that have contributed to the research in the pharmacol-ogy of lifespan extension and the understanding of the aging process are discussed. Results: Dietary restriction (DR) constitutes a common experimental process to extend life span in all organisms. Besides, classical antiaging drugs such as resveratrol, rapamycin and metformin denominated as DR mimetics are also discussed. Likewise, the main therapeutic targets of these drugs include sirtuins, IGF-1, and mTOR, all of them being modulated by DR. Conclusion: Advances in molecular biology have uncovered the potential molecular pathways involved in the aging process. Due to their characteristics, invertebrate models are mainly used for drug screening. The National Institute on Aging (NIA) developed the Interventions Testing Program (ITP). At the pre-clinical level, the ITP uses Heterogeneous mouse model (HET) which is probably the most suitable rodent model to study potential drugs against aging prevention. The accelerated-senescence mouse P8 is also a mammalian rodent model for aging research. However, when evaluating the effect of drugs on a preclinical level, the evaluation must be done in non-human primates since it is the mammalian specie closest to humans. Research is needed to investigate the impact of new potential drugs for the increase of human quality of
Collapse
Affiliation(s)
- Jaume Folch
- Unitat de Bioquimica i Biotecnologia, Facultat de Medicina i Ciencies de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Oriol Busquets
- Unitat de Bioquimica i Biotecnologia, Facultat de Medicina i Ciencies de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Departament Deaprtament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Miren Ettcheto
- Unitat de Bioquimica i Biotecnologia, Facultat de Medicina i Ciencies de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Departament Deaprtament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Elena Sánchez-López
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Unitat de Farmacia, Tecnologia Farmacèutica i Fisico-química, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Mercè Pallàs
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Departament Deaprtament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Carlos Beas-Zarate
- Departamento de Biologia Celulary Molecular, C.U.C.B.A., Universidad de Guadalajara and Division de Neurociencias, Sierra Mojada 800, Col. Independencia, Guadalajara, Jalisco 44340, Mexico
| | - Miguel Marin
- Centro de Biotecnologia. Universidad Nacional de Loja, Av. Pío Jaramillo Alvarado y Reinaldo Espinosa, La Argelia. Loja, Ecuador
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Jordi Olloquequi
- Instituto de Ciencias Biomedicas, Facultad de Ciencias de la Salud, Universidad Autonoma de Chile, Talca, Chile
| | - Carme Auladell
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Departamento de Biologia Celulary Molecular, C.U.C.B.A., Universidad de Guadalajara and Division de Neurociencias, Sierra Mojada 800, Col. Independencia, Guadalajara, Jalisco 44340, Mexico.,Departament de Biologia Cellular, Fisiologia i Inmunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Antoni Camins
- Departament Deaprtament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Centro de Biotecnologia. Universidad Nacional de Loja, Av. Pío Jaramillo Alvarado y Reinaldo Espinosa, La Argelia. Loja, Ecuador
| |
Collapse
|
31
|
López-Lluch G, Hernández-Camacho JD, Fernández-Ayala DJM, Navas P. Mitochondrial dysfunction in metabolism and ageing: shared mechanisms and outcomes? Biogerontology 2018; 19:461-480. [PMID: 30143941 DOI: 10.1007/s10522-018-9768-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/21/2018] [Indexed: 12/15/2022]
Abstract
Mitochondria are key in the metabolism of aerobic organisms and in ageing progression and age-related diseases. Mitochondria are essential for obtaining ATP from glucose and fatty acids but also in many other essential functions in cells including aminoacids metabolism, pyridine synthesis, phospholipid modifications and calcium regulation. On the other hand, the activity of mitochondria is also the principal source of reactive oxygen species in cells. Ageing and chronic age-related diseases are associated with the deregulation of cell metabolism and dysfunction of mitochondria. Cell metabolism is controlled by three major nutritional sensors: mTOR, AMPK and Sirtuins. These factors control mitochondrial biogenesis and dynamics by regulating fusion, fission and turnover through mito- and autophagy. A complex interaction between the activity of these nutritional sensors, mitochondrial biogenesis rate and dynamics exists and affect ageing, age-related diseases including metabolic disease. Further, mitochondria maintain a constant communication with nucleus modulating gene expression and modifying epigenetics. In this review we highlight the importance of mitochondria in ageing and the repercussion in the progression of age-related diseases and metabolic disease.
Collapse
Affiliation(s)
- Guillermo López-Lluch
- Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide, Carretera de Utrera km. 1, 41013, Seville, Spain.
| | - Juan Diego Hernández-Camacho
- Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide, Carretera de Utrera km. 1, 41013, Seville, Spain
| | - Daniel J Moreno Fernández-Ayala
- Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide, Carretera de Utrera km. 1, 41013, Seville, Spain
| | - Plácido Navas
- Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide, Carretera de Utrera km. 1, 41013, Seville, Spain
| |
Collapse
|
32
|
McMurphy T, Huang W, Queen NJ, Ali S, Widstrom KJ, Liu X, Xiao R, Siu JJ, Cao L. Implementation of environmental enrichment after middle age promotes healthy aging. Aging (Albany NY) 2018; 10:1698-1721. [PMID: 30036185 PMCID: PMC6075449 DOI: 10.18632/aging.101502] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 07/15/2018] [Indexed: 12/19/2022]
Abstract
With increases in life expectancy, it is vital to understand the dynamics of aging, their interaction with lifestyle factors, and the connections to age-related disease processes. Our work on environmental enrichment (EE), a housing environment boosting mental health, has revealed a novel anticancer and anti-obesity phenotype mediated by a brain-fat axis: the hypothalamic-sympathoneural-adipocyte (HSA) axis in young animals. Here we investigated EE effects on healthspan and lifespan when initiated after middle age. Short-term EE for six weeks activated the HSA axis in 10-month-old mice. Long-term EE for twelve months reduced adiposity, improved glucose tolerance, decreased leptin levels, enhanced motor abilities, and inhibited anxiety. In addition to adipose remodeling, EE decreased age-related liver steatosis, reduced hepatic glucose production, and increased glucose uptake by liver and adipose tissue contributing to the improved glycemic control. The EE-induced liver modulation was associated with a suppression of protein kinase Cε. Moreover, EE down-regulated the expression of inflammatory genes in the brain, adipose, and liver. EE initiated at 18-month of age significantly improved glycemic control and showed a trend of positive impact on mean lifespan. These data suggest that EE induces metabolic and behavioral adaptations that are shared by factors known to increase healthspan and lifespan.
Collapse
Affiliation(s)
- Travis McMurphy
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Equal contribution
| | - Wei Huang
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Equal contribution
| | - Nicholas J. Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Seemaab Ali
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kyle J. Widstrom
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Xianglan Liu
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Run Xiao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jason J. Siu
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
33
|
Siddiqui S, Lustig A, Carter A, Sankar M, Daimon CM, Premont RT, Etienne H, van Gastel J, Azmi A, Janssens J, Becker KG, Zhang Y, Wood W, Lehrmann E, Martin JG, Martin B, Taub DD, Maudsley S. Genomic deletion of GIT2 induces a premature age-related thymic dysfunction and systemic immune system disruption. Aging (Albany NY) 2017; 9:706-740. [PMID: 28260693 PMCID: PMC5391227 DOI: 10.18632/aging.101185] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 02/19/2017] [Indexed: 12/12/2022]
Abstract
Recent research has proposed that GIT2 (G protein-coupled receptor kinase interacting protein 2) acts as an integrator of the aging process through regulation of 'neurometabolic' integrity. One of the commonly accepted hallmarks of the aging process is thymic involution. At a relatively young age, 12 months old, GIT2-/- mice present a prematurely distorted thymic structure and dysfunction compared to age-matched 12 month-old wild-type control (C57BL/6) mice. Disruption of thymic structure in GIT2-/- (GIT2KO) mice was associated with a significant reduction in the expression of the cortical thymic marker, Troma-I (cytokeratin 8). Double positive (CD4+CD8+) and single positive CD4+ T cells were also markedly reduced in 12 month-old GIT2KO mice compared to age-matched control wild-type mice. Coincident with this premature thymic disruption in GIT2KO mice was the unique generation of a novel cervical 'organ', i.e. 'parathymic lobes'. These novel organs did not exhibit classical peripheral lymph node-like characteristics but expressed high levels of T cell progenitors that were reflexively reduced in GIT2KO thymi. Using signaling pathway analysis of GIT2KO thymus and parathymic lobe transcriptomic data we found that the molecular signaling functions lost in the dysfunctional GIT2KO thymus were selectively reinstated in the novel parathymic lobe - suggestive of a compensatory effect for the premature thymic disruption. Broader inspection of high-dimensionality transcriptomic data from GIT2KO lymph nodes, spleen, thymus and parathymic lobes revealed a systemic alteration of multiple proteins (Dbp, Tef, Per1, Per2, Fbxl3, Ddit4, Sin3a) involved in the multidimensional control of cell cycle clock regulation, cell senescence, cellular metabolism and DNA damage. Altered cell clock regulation across both immune and non-immune tissues therefore may be responsible for the premature 'aging' phenotype of GIT2KO mice.
Collapse
Affiliation(s)
- Sana Siddiqui
- Receptor Pharmacology Unit, Laboratory of Neurosciences, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD 21224, USA
| | - Ana Lustig
- Laboratory of Molecular Biology and Immunology, NIA, NIH, Baltimore, MD 21224, USA
| | - Arnell Carter
- Laboratory of Molecular Biology and Immunology, NIA, NIH, Baltimore, MD 21224, USA
| | - Mathavi Sankar
- Metabolism Unit, Laboratory of Clinical Investigation, NIA, NIH, Baltimore, MD 21224, USA
| | - Caitlin M Daimon
- Metabolism Unit, Laboratory of Clinical Investigation, NIA, NIH, Baltimore, MD 21224, USA
| | | | - Harmonie Etienne
- Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, Belgium
| | - Jaana van Gastel
- Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, Belgium
| | - Abdelkrim Azmi
- Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, Belgium
| | - Jonathan Janssens
- Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, Belgium
| | - Kevin G Becker
- Gene Expression and Genomics Unit, Research Resources Branch, NIA, NIH, Baltimore, MD 21224, USA
| | - Yongqing Zhang
- Gene Expression and Genomics Unit, Research Resources Branch, NIA, NIH, Baltimore, MD 21224, USA
| | - William Wood
- Gene Expression and Genomics Unit, Research Resources Branch, NIA, NIH, Baltimore, MD 21224, USA
| | - Elin Lehrmann
- Gene Expression and Genomics Unit, Research Resources Branch, NIA, NIH, Baltimore, MD 21224, USA
| | - James G Martin
- Research Institute of the MUHC, Centre for Translational Biology (CTB), Meakins-Christie Laboratories, McGill University, Montreal, QC, H4A 3J1, Canada
| | - Bronwen Martin
- Metabolism Unit, Laboratory of Clinical Investigation, NIA, NIH, Baltimore, MD 21224, USA
| | - Dennis D Taub
- Laboratory of Molecular Biology and Immunology, NIA, NIH, Baltimore, MD 21224, USA
| | - Stuart Maudsley
- Receptor Pharmacology Unit, Laboratory of Neurosciences, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD 21224, USA.,Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, Belgium
| |
Collapse
|
34
|
Merry TL, Kuhlow D, Laube B, Pöhlmann D, Pfeiffer AFH, Kahn CR, Ristow M, Zarse K. Impairment of insulin signalling in peripheral tissue fails to extend murine lifespan. Aging Cell 2017; 16:761-772. [PMID: 28544360 PMCID: PMC5506415 DOI: 10.1111/acel.12610] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2017] [Indexed: 01/02/2023] Open
Abstract
Impaired insulin/IGF1 signalling has been shown to extend lifespan in model organisms ranging from yeast to mammals. Here we sought to determine the effect of targeted disruption of the insulin receptor (IR) in non‐neuronal tissues of adult mice on the lifespan. We induced hemizygous (PerIRKO+/−) or homozygous (PerIRKO−/−) disruption of the IR in peripheral tissue of 15‐weeks‐old mice using a tamoxifen‐inducible Cre transgenic mouse with only peripheral tissue expression, and subsequently monitored glucose metabolism, insulin signalling and spontaneous death rates over 4 years. Complete peripheral IR disruption resulted in a diabetic phenotype with increased blood glucose and plasma insulin levels in young mice. Although blood glucose levels returned to normal, and fat mass was reduced in aged PerIRKO−/− mice, their lifespan was reduced. By contrast, heterozygous disruption had no effect on lifespan. This was despite young male PerIRKO+/− mice showing reduced fat mass and mild increase in hepatic insulin sensitivity. In conflict with findings in metazoans like Caenorhabditis elegans and Drosophila melanogaster, our results suggest that heterozygous impairment of the insulin signalling limited to peripheral tissues of adult mice fails to extend lifespan despite increased systemic insulin sensitivity, while homozygous impairment shortens lifespan.
Collapse
Affiliation(s)
- Troy L. Merry
- Energy Metabolism Laboratory; Swiss Federal Institute of Technology (ETH) Zurich-Schwerzenbach; Schwerzenbach, Zurich Switzerland
- School of Medical Sciences; University of Auckland; Auckland New Zealand
| | - Doreen Kuhlow
- Department of Human Nutrition; Friedrich Schiller-University; Jena Germany
- Department of Human Nutrition; German Institute of Human Nutrition Potsdam-Rehbrücke; Nuthetal Germany
| | - Beate Laube
- Energy Metabolism Laboratory; Swiss Federal Institute of Technology (ETH) Zurich-Schwerzenbach; Schwerzenbach, Zurich Switzerland
- Department of Human Nutrition; Friedrich Schiller-University; Jena Germany
| | - Doris Pöhlmann
- Energy Metabolism Laboratory; Swiss Federal Institute of Technology (ETH) Zurich-Schwerzenbach; Schwerzenbach, Zurich Switzerland
| | - Andreas F. H. Pfeiffer
- Department of Human Nutrition; German Institute of Human Nutrition Potsdam-Rehbrücke; Nuthetal Germany
- Medizinische Klinik für Endokrinologie; Diabetes und Ernährungsmedizin; Charité University Medicine Berlin; Berlin Germany
| | - C. Ronald Kahn
- Section on Integrative Physiology and Metabolism; Joslin Diabetes Center and Department of Medicine; Boston MA USA
| | - Michael Ristow
- Energy Metabolism Laboratory; Swiss Federal Institute of Technology (ETH) Zurich-Schwerzenbach; Schwerzenbach, Zurich Switzerland
- Department of Human Nutrition; Friedrich Schiller-University; Jena Germany
- Department of Human Nutrition; German Institute of Human Nutrition Potsdam-Rehbrücke; Nuthetal Germany
| | - Kim Zarse
- Energy Metabolism Laboratory; Swiss Federal Institute of Technology (ETH) Zurich-Schwerzenbach; Schwerzenbach, Zurich Switzerland
- Department of Human Nutrition; Friedrich Schiller-University; Jena Germany
| |
Collapse
|
35
|
Xu J, Wang L, Tang J, Jia G, Liu G, Chen X, Cai J, Shang H, Zhao H. Pancreatic atrophy caused by dietary selenium deficiency induces hypoinsulinemic hyperglycemia via global down-regulation of selenoprotein encoding genes in broilers. PLoS One 2017; 12:e0182079. [PMID: 28763492 PMCID: PMC5538751 DOI: 10.1371/journal.pone.0182079] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/05/2017] [Indexed: 12/15/2022] Open
Abstract
This study was envisaged to comprehensively profile genes in selected tissues along with a few biochemical indicators and integrate resulting information with dietary selenium (Se) deficiency symptoms in broilers. A total of 120 one-day-old Cobb male broilers were equally divided into two groups and fed a Se deficient corn-soybean-based basal diet supplemented with 0.3 mg/kg sodium selenite (Control, Se adequate) or without selenite (Se deficiency) for five weeks. Effects of Se deficiency on mRNA abundance of twenty-three selenoprotein encoding genes and seventeen insulin signaling related genes were studied at day 35 in pancreas, liver and muscle along with plasma biochemical constituents and enzyme activities. Compared to healthy birds in control diet, Se deficient diet induced deficiency symptoms in 90% birds and classic nutritional pancreatic atrophy, depressed growth performance of broilers, and decreased (P < 0.01 to P < 0.05) total antioxidant capacity and activities of superoxide dismutase and glutathione peroxidase in plasma and three other tissues. Se deficiency resulted in 58% higher mortality than control birds. Dietary Se deficiency down-regulated (P < 0.01-0.05) eighteen selenoprotein encoding genes in pancreas, fourteen genes in muscle and nine genes in liver, and up-regulated (P < 0.05) Txnrd1 and Selx in liver. Meanwhile, six, thirteen and five insulin signaling related genes were down-regulated (P < 0.01-0.05) in pancreas, muscle and liver, respectively, and three genes were up-regulated (P < 0.01) in liver. The decrease (P < 0.05) in levels of plasma insulin, total triglyceride and total cholesterol, and concurrent elevated (P < 0.05) levels of plasma glucose and inflammatory cytokines accompanied the global down-regulation of selenoprotein encoding- and insulin signaling related- genes in Se deficient birds. It was concluded that dietary Se deficiency induces nutritional pancreatic atrophy and metabolic disorder of glucose and lipid in broilers via down-regulation of selenoprotein encoding- and insulin signaling related- genes, indicating potential roles of these genes in metabolic regulation.
Collapse
Affiliation(s)
- Jingyang Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Longqiong Wang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jiayong Tang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Trace Element Research Center, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Gang Jia
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Trace Element Research Center, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Guangmang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaoling Chen
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jingyi Cai
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Haiying Shang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hua Zhao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Trace Element Research Center, Sichuan Agricultural University, Chengdu, Sichuan, China
- * E-mail:
| |
Collapse
|
36
|
Tajima K, Shirakawa J, Togashi Y, Yamazaki S, Okuyama T, Kyohara M, Konishi H, Terauchi Y. Metabolic recovery of lipodystrophy, liver steatosis, and pancreatic β cell proliferation after the withdrawal of OSI-906. Sci Rep 2017. [PMID: 28646158 PMCID: PMC5482874 DOI: 10.1038/s41598-017-04304-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Growth factor signaling via insulin receptor (IR) and IGF-1 receptor (IGF1R) plays several important roles in the pathogenesis of metabolic syndrome and diabetes. OSI-906 (linsitinib), an anti-tumor drug, is an orally bioavailable dual inhibitor of IR and IGF1R. To investigate the recovery from metabolic changes induced by the acute inhibition of IR and IGF1R in adult mice, mice were treated with OSI-906 or a vehicle for 7 days and the results were analyzed on the last day of injection (Day 7) or after 7 or 21 days of withdrawal (Day 14 or Day 28). On day 7, the visceral white fat mass was significantly reduced in mice treated with OSI-906 accompanied by a reduced expression of leptin and an increased expression of the lipolysis-related genes Lpl and Atgl. Interestingly, the lipoatrophy and the observed changes in gene expression were completely reversed on day 14. Similarly, liver steatosis and β cell proliferation were transiently observed on day 7 but had disappeared by day 14. Taken together, these results suggest that this model for the acute inhibition of systemic IR/IGF1R signaling may be useful for investigating the recovery from metabolic disorders induced by impaired growth factor signaling.
Collapse
Affiliation(s)
- Kazuki Tajima
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan
| | - Jun Shirakawa
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan.
| | - Yu Togashi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan
| | - Shunsuke Yamazaki
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan
| | - Tomoko Okuyama
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan
| | - Mayu Kyohara
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan
| | - Hiromi Konishi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan, 236-0004, Japan.
| |
Collapse
|
37
|
Boengler K, Kosiol M, Mayr M, Schulz R, Rohrbach S. Mitochondria and ageing: role in heart, skeletal muscle and adipose tissue. J Cachexia Sarcopenia Muscle 2017; 8:349-369. [PMID: 28432755 PMCID: PMC5476857 DOI: 10.1002/jcsm.12178] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/23/2016] [Accepted: 11/24/2016] [Indexed: 12/11/2022] Open
Abstract
Age is the most important risk factor for most diseases. Mitochondria play a central role in bioenergetics and metabolism. In addition, several lines of evidence indicate the impact of mitochondria in lifespan determination and ageing. The best-known hypothesis to explain ageing is the free radical theory, which proposes that cells, organs, and organisms age because they accumulate reactive oxygen species (ROS) damage over time. Mitochondria play a central role as the principle source of intracellular ROS, which are mainly formed at the level of complex I and III of the respiratory chain. Dysfunctional mitochondria generating less ATP have been observed in various aged organs. Mitochondrial dysfunction comprises different features including reduced mitochondrial content, altered mitochondrial morphology, reduced activity of the complexes of the electron transport chain, opening of the mitochondrial permeability transition pore, and increased ROS formation. Furthermore, abnormalities in mitochondrial quality control or defects in mitochondrial dynamics have also been linked to senescence. Among the tissues affected by mitochondrial dysfunction are those with a high-energy demand and thus high mitochondrial content. Therefore, the present review focuses on the impact of mitochondria in the ageing process of heart and skeletal muscle. In this article, we review different aspects of mitochondrial dysfunction and discuss potential therapeutic strategies to improve mitochondrial function. Finally, novel aspects of adipose tissue biology and their involvement in the ageing process are discussed.
Collapse
Affiliation(s)
- Kerstin Boengler
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| | - Maik Kosiol
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| | - Susanne Rohrbach
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| |
Collapse
|
38
|
Fujii N, Narita T, Okita N, Kobayashi M, Furuta Y, Chujo Y, Sakai M, Yamada A, Takeda K, Konishi T, Sudo Y, Shimokawa I, Higami Y. Sterol regulatory element-binding protein-1c orchestrates metabolic remodeling of white adipose tissue by caloric restriction. Aging Cell 2017; 16:508-517. [PMID: 28256090 PMCID: PMC5418191 DOI: 10.1111/acel.12576] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2017] [Indexed: 12/31/2022] Open
Abstract
Caloric restriction (CR) can delay onset of several age‐related pathophysiologies and extend lifespan in various species, including rodents. CR also induces metabolic remodeling involved in activation of lipid metabolism, enhancement of mitochondrial biogenesis, and reduction of oxidative stress in white adipose tissue (WAT). In studies using genetically modified mice with extended lifespans, WAT characteristics influenced mammalian lifespans. However, molecular mechanisms underlying CR‐associated metabolic remodeling of WAT remain unclear. Sterol regulatory element‐binding protein‐1c (Srebp‐1c), a master transcription factor of fatty acid (FA) biosynthesis, is responsible for the pathogenesis of fatty liver (steatosis). Our study showed that, under CR conditions, Srebp‐1c enhanced mitochondrial biogenesis via increased expression of peroxisome proliferator‐activated receptor gamma coactivator‐1α (Pgc‐1α) and upregulated expression of proteins involved in FA biosynthesis within WAT. However, via Srebp‐1c, most of these CR‐associated metabolic alterations were not observed in other tissues, including the liver. Moreover, our data indicated that Srebp‐1c may be an important factor both for CR‐associated suppression of oxidative stress, through increased synthesis of glutathione in WAT, and for the prolongevity action of CR. Our results strongly suggested that Srebp‐1c, the primary FA biosynthesis‐promoting transcriptional factor implicated in fatty liver disease, is also the food shortage‐responsive factor in WAT. This indicated that Srebp‐1c is a key regulator of metabolic remodeling leading to the beneficial effects of CR.
Collapse
Affiliation(s)
- Namiki Fujii
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Takumi Narita
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Naoyuki Okita
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Department of Internal Medicine Research; Sasaki Institute; Sasaki Foundation; 2-2 Kandasurugadai Chiyoda-ku, Tokyo 101-0062 Japan
| | - Masaki Kobayashi
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Yurika Furuta
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Yoshikazu Chujo
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Masahiro Sakai
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Atsushi Yamada
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Kanae Takeda
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Tomokazu Konishi
- Faculty of Bioresource Sciences; Akita Prefectural University; Shimoshinjo Nakano, Akita 010-0195 Japan
| | - Yuka Sudo
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Isao Shimokawa
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Department of Pathology; Nagasaki University Graduate School of Biomedical Sciences; 1-12-4 Sakamoto Nagasaki 852-8523 Japan
| | - Yoshikazu Higami
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| |
Collapse
|
39
|
Esparza-Moltó PB, Nuevo-Tapioles C, Cuezva JM. Regulation of the H +-ATP synthase by IF1: a role in mitohormesis. Cell Mol Life Sci 2017; 74:2151-2166. [PMID: 28168445 PMCID: PMC5425498 DOI: 10.1007/s00018-017-2462-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 01/18/2023]
Abstract
The mitochondrial H+-ATP synthase is a primary hub of cellular homeostasis by providing the energy required to sustain cellular activity and regulating the production of signaling molecules that reprogram nuclear activity needed for adaption to changing cues. Herein, we summarize findings regarding the regulation of the activity of the H+-ATP synthase by its physiological inhibitor, the ATPase inhibitory factor 1 (IF1) and their functional role in cellular homeostasis. First, we outline the structure and the main molecular mechanisms that regulate the activity of the enzyme. Next, we describe the molecular biology of IF1 and summarize the regulation of IF1 expression and activity as an inhibitor of the H+-ATP synthase emphasizing the role of IF1 as a main driver of energy rewiring and cellular signaling in cancer. Findings in transgenic mice in vivo indicate that the overexpression of IF1 is sufficient to reprogram energy metabolism to an enhanced glycolysis and activate reactive oxygen species (ROS)-dependent signaling pathways that promote cell survival. These findings are placed in the context of mitohormesis, a program in which a mild mitochondrial stress triggers adaptive cytoprotective mechanisms that improve lifespan. In this regard, we emphasize the role played by the H+-ATP synthase in modulating signaling pathways that activate the mitohormetic response, namely ATP, ROS and target of rapamycin (TOR). Overall, we aim to highlight the relevant role of the H+-ATP synthase and of IF1 in cellular physiology and the need of additional studies to decipher their contributions to aging and age-related diseases.
Collapse
Affiliation(s)
- Pau B Esparza-Moltó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Cristina Nuevo-Tapioles
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28049, Madrid, Spain.
| |
Collapse
|
40
|
Templeman NM, Skovsø S, Page MM, Lim GE, Johnson JD. A causal role for hyperinsulinemia in obesity. J Endocrinol 2017; 232:R173-R183. [PMID: 28052999 DOI: 10.1530/joe-16-0449] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 01/03/2017] [Indexed: 12/13/2022]
Abstract
Insulin modulates the biochemical pathways controlling lipid uptake, lipolysis and lipogenesis at multiple levels. Elevated insulin levels are associated with obesity, and conversely, dietary and pharmacological manipulations that reduce insulin have occasionally been reported to cause weight loss. However, the causal role of insulin hypersecretion in the development of mammalian obesity remained controversial in the absence of direct loss-of-function experiments. Here, we discuss theoretical considerations around the causal role of excess insulin for obesity, as well as recent studies employing mice that are genetically incapable of the rapid and sustained hyperinsulinemia that normally accompanies a high-fat diet. We also discuss new evidence demonstrating that modest reductions in circulating insulin prevent weight gain, with sustained effects that can persist after insulin levels normalize. Importantly, evidence from long-term studies reveals that a modest reduction in circulating insulin is not associated with impaired glucose homeostasis, meaning that body weight and lipid homeostasis are actually more sensitive to small changes in circulating insulin than glucose homeostasis in these models. Collectively, the evidence from new studies on genetic loss-of-function models forces a re-evaluation of current paradigms related to obesity, insulin resistance and diabetes. The potential for translation of these findings to humans is briefly discussed.
Collapse
Affiliation(s)
- Nicole M Templeman
- Department of Cellular and Physiological SciencesDiabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Søs Skovsø
- Department of Cellular and Physiological SciencesDiabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Melissa M Page
- Department of Cellular and Physiological SciencesDiabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gareth E Lim
- Department of Cellular and Physiological SciencesDiabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - James D Johnson
- Department of Cellular and Physiological SciencesDiabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Institute for Personalized Therapeutic NutritionVancouver, British Columbia, Canada
| |
Collapse
|
41
|
DJ-1 maintains energy and glucose homeostasis by regulating the function of brown adipose tissue. Cell Discov 2017; 3:16054. [PMID: 28224045 PMCID: PMC5309696 DOI: 10.1038/celldisc.2016.54] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 12/28/2016] [Indexed: 12/12/2022] Open
Abstract
DJ-1 protein is involved in multiple physiological processes, including Parkinson’s disease. However, the role of DJ-1 in the metabolism is largely unknown. Here we found that DJ-1 maintained energy balance and glucose homeostasisvia regulating brown adipose tissue (BAT) activity. DJ-1-deficient mice reduced body mass, increased energy expenditure and improved insulin sensitivity. DJ-1 deletion also resisted high-fat-diet (HFD) induced obesity and insulin resistance. Accordingly, DJ-1 transgene triggered autonomous obesity and glucose intolerance. Further BAT transplantation experiments clarified DJ-1 regulates energy and glucose homeostasis by modulating BAT function. Mechanistically, we found that DJ-1 promoted PTEN proteasomal degradation via an E3 ligase, mind bomb-2 (Mib2), which led to Akt activation and inhibited FoxO1-dependent Ucp1 (Uncoupling protein-1) expression in BAT. Consistently, ablation of Akt1 mitigated the obesity and BAT dysfunction induced by DJ-1 transgene. These findings define a new biological role of DJ-1 protein in regulating BAT function, with an implication of the therapeutic target in the treatment of metabolic disorders.
Collapse
|
42
|
Different Mechanisms of Longevity in Long-Lived Mouse and Caenorhabditis elegans Mutants Revealed by Statistical Analysis of Mortality Rates. Genetics 2016; 204:905-920. [PMID: 27638422 DOI: 10.1534/genetics.116.192369] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/28/2016] [Indexed: 12/23/2022] Open
Abstract
Mouse and Caenorhabditis elegans mutants with altered life spans are being used to investigate the aging process and how genes determine life span. The survival of a population can be modeled by the Gompertz function, which comprises two parameters. One of these parameters ("G") describes the rate at which mortality accelerates with age and is often described as the "rate of aging." The other parameter ("A") may correspond to the organism's baseline vulnerability to deleterious effects of disease and the environment. We show that, in mice, life-span-extending mutations systematically fail to affect the age-dependent acceleration of mortality (G), but instead affect only baseline vulnerability (A). This remains true even when comparing strains maintained under identical environmental conditions. In contrast, life-span-extending mutations in C. elegans were associated with decreases in G These observations on mortality rate kinetics suggest that the mechanisms of aging in mammals might fundamentally differ from those in nematodes.
Collapse
|
43
|
Abstract
Hormesis is a process whereby exposure to a low dose of a potentially harmful stressor promotes adaptive changes to the cell that enables it to better tolerate subsequent stress. In recent years this concept has been applied specifically to the mitochondria (mitohormesis), suggesting that in response to a perturbation the mitochondria can initiate and transduce a signal to the nucleus that coordinates a transcriptional response resulting in both mitochondrial and non-mitochondrial adaptations that return and maintain cellular homeostasis. In this review we summarize the evidence that mitohormesis is a significant adaptive-response signaling pathway, and suggest that it plays a role in mediating exercise-induced adaptations. We discuss potential mitochondrial emitters of retrograde signals that may activate known exercise-sensitive transcription factors to modulate transcription responses to exercise, and draw on evidence from mitochondrial dysfunction animal models to support a role for mitohormesis in mitochondrial biogenesis. Studies directly linking mitohormesis to the exercise training response are lacking, however mounting evidence suggests numerous signals are emitted from the mitochondria during exercise and have the potential to induce a nuclear transcription response, with reactive oxygen species (ROS) being the primary candidate.
Collapse
Affiliation(s)
- Troy L Merry
- Energy Metabolism Laboratory, Swiss Federal Institute of Technology (ETH), 8603 Zurich, Switzerland.
| | - Michael Ristow
- Energy Metabolism Laboratory, Swiss Federal Institute of Technology (ETH), 8603 Zurich, Switzerland
| |
Collapse
|
44
|
MacPherson REK, Dragos SM, Ramos S, Sutton C, Frendo-Cumbo S, Castellani L, Watt MJ, Perry CGR, Mutch DM, Wright DC. Reduced ATGL-mediated lipolysis attenuates β-adrenergic-induced AMPK signaling, but not the induction of PKA-targeted genes, in adipocytes and adipose tissue. Am J Physiol Cell Physiol 2016; 311:C269-76. [PMID: 27357546 DOI: 10.1152/ajpcell.00126.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/23/2016] [Indexed: 12/25/2022]
Abstract
5'-AMP-activated protein kinase (AMPK) is activated as a consequence of lipolysis and has been shown to play a role in regulation of adipose tissue mitochondrial content. Conversely, the inhibition of lipolysis has been reported to potentiate the induction of protein kinase A (PKA)-targeted genes involved in the regulation of oxidative metabolism. The purpose of the current study was to address these apparent discrepancies and to more fully examine the relationship between lipolysis, AMPK, and the β-adrenergic-mediated regulation of gene expression. In 3T3-L1 adipocytes, the adipose tissue triglyceride lipase (ATGL) inhibitor ATGListatin attenuated the Thr(172) phosphorylation of AMPK by a β3-adrenergic agonist (CL 316,243) independent of changes in PKA signaling. Similarly, CL 316,243-induced increases in the Thr(172) phosphorylation of AMPK were reduced in adipose tissue from whole body ATGL-deficient mice. Despite reductions in the activation of AMPK, the induction of PKA-targeted genes was intact or, in some cases, increased. Similarly, markers of mitochondrial content and respiration were increased in adipose tissue from ATGL knockout mice independent of changes in the Thr(172) phosphorylation of AMPK. Taken together, our data provide evidence that AMPK is not required for the regulation of adipose tissue oxidative capacity in conditions of reduced fatty acid release.
Collapse
Affiliation(s)
- Rebecca E K MacPherson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Steven M Dragos
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Sofhia Ramos
- School of Kinesiology and Health Sciences, York University, Toronto, Ontario, Canada; and
| | - Charles Sutton
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Scott Frendo-Cumbo
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Laura Castellani
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Matthew J Watt
- Monash Biomedicine Discovery Institute, Metabolic Disease and Obesity Program, and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Christopher G R Perry
- School of Kinesiology and Health Sciences, York University, Toronto, Ontario, Canada; and
| | - David M Mutch
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - David C Wright
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada;
| |
Collapse
|
45
|
Aging and adipose tissue: potential interventions for diabetes and regenerative medicine. Exp Gerontol 2016; 86:97-105. [PMID: 26924669 DOI: 10.1016/j.exger.2016.02.013] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/22/2016] [Accepted: 02/24/2016] [Indexed: 12/15/2022]
Abstract
Adipose tissue dysfunction occurs with aging and has systemic effects, including peripheral insulin resistance, ectopic lipid deposition, and inflammation. Fundamental aging mechanisms, including cellular senescence and progenitor cell dysfunction, occur in adipose tissue with aging and may serve as potential therapeutic targets in age-related disease. In this review, we examine the role of adipose tissue in healthy individuals and explore how aging leads to adipose tissue dysfunction, redistribution, and changes in gene regulation. Adipose tissue plays a central role in longevity, and interventions restricted to adipose tissue may impact lifespan. Conversely, obesity may represent a state of accelerated aging. We discuss the potential therapeutic potential of targeting basic aging mechanisms, including cellular senescence, in adipose tissue, using type II diabetes and regenerative medicine as examples. We make the case that aging should not be neglected in the study of adipose-derived stem cells for regenerative medicine strategies, as elderly patients make up a large portion of individuals in need of such therapies.
Collapse
|
46
|
Haussler MR, Whitfield GK, Haussler CA, Sabir MS, Khan Z, Sandoval R, Jurutka PW. 1,25-Dihydroxyvitamin D and Klotho: A Tale of Two Renal Hormones Coming of Age. VITAMINS AND HORMONES 2016; 100:165-230. [PMID: 26827953 DOI: 10.1016/bs.vh.2015.11.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
1,25-Dihydroxyvitamin D3 (1,25D) is the renal metabolite of vitamin D that signals through binding to the nuclear vitamin D receptor (VDR). The ligand-receptor complex transcriptionally regulates genes encoding factors stimulating calcium and phosphate absorption plus bone remodeling, maintaining a skeleton with reduced risk of age-related osteoporotic fractures. 1,25D/VDR signaling exerts feedback control of Ca/PO4 via regulation of FGF23, klotho, and CYP24A1 to prevent age-related, ectopic calcification, fibrosis, and associated pathologies. Vitamin D also elicits xenobiotic detoxification, oxidative stress reduction, neuroprotective functions, antimicrobial defense, immunoregulation, anti-inflammatory/anticancer actions, and cardiovascular benefits. Many of the healthspan advantages conferred by 1,25D are promulgated by its induction of klotho, a renal hormone that is an anti-aging enzyme/coreceptor that protects against skin atrophy, osteopenia, hyperphosphatemia, endothelial dysfunction, cognitive defects, neurodegenerative disorders, and impaired hearing. In addition to the high-affinity 1,25D hormone, low-affinity nutritional VDR ligands including curcumin, polyunsaturated fatty acids, and anthocyanidins initiate VDR signaling, whereas the longevity principles resveratrol and SIRT1 potentiate VDR signaling. 1,25D exerts actions against neural excitotoxicity and induces serotonin mood elevation to support cognitive function and prosocial behavior. Together, 1,25D and klotho maintain the molecular signaling systems that promote growth (p21), development (Wnt), antioxidation (Nrf2/FOXO), and homeostasis (FGF23) in tissues crucial for normal physiology, while simultaneously guarding against malignancy and degeneration. Therefore, liganded-VDR modulates the expression of a "fountain of youth" array of genes, with the klotho target emerging as a major player in the facilitation of health span by delaying the chronic diseases of aging.
Collapse
Affiliation(s)
- Mark R Haussler
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, USA.
| | - G Kerr Whitfield
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Carol A Haussler
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Marya S Sabir
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, Arizona, USA
| | - Zainab Khan
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, Arizona, USA
| | - Ruby Sandoval
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, Arizona, USA
| | - Peter W Jurutka
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, USA; School of Mathematical and Natural Sciences, Arizona State University, Glendale, Arizona, USA
| |
Collapse
|
47
|
Templeman NM, Clee SM, Johnson JD. Suppression of hyperinsulinaemia in growing female mice provides long-term protection against obesity. Diabetologia 2015; 58:2392-402. [PMID: 26155745 PMCID: PMC4572061 DOI: 10.1007/s00125-015-3676-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/05/2015] [Indexed: 01/13/2023]
Abstract
AIMS/HYPOTHESIS Hyperinsulinaemia is associated with obesity but its causal role in the onset of obesity remains controversial. In this study, we tested the hypothesis that transient attenuation of diet-induced insulin hypersecretion in young mice can provide sustained protection against obesity throughout adult life. METHODS Using 'genetically humanised' mice lacking both alleles of rodent-specific Ins1, we compared mice heterozygous for the ancestral insulin gene Ins2 with Ins2(+/+) controls. Female Ins1(-/-):Ins2(+/-) and Ins1(-/-):Ins2(+/+) littermates were fed chow or high-fat diet (HFD). Insulin secretion, metabolic health variables and body mass/composition were tracked for over 1 year. We examined islet function and adipose transcript levels of adipogenic, lipogenic and lipolytic genes at two time points. RESULTS In control Ins1(-/-):Ins2(+/+) mice, HFD resulted in elevated fasting and glucose-stimulated insulin secretion between 8 weeks and 27 weeks of age. Hyperinsulinaemia was reduced by nearly 50% in Ins1(-/-):Ins2(+/-) mice during this period, without lasting adverse effects on glucose homeostasis. This corresponded with attenuated weight gain and adiposity. White adipose tissue from Ins1(-/-):Ins2(+/-) mice had fewer large lipid droplets, although transcriptional changes were not detected. Importantly, Ins1(-/-):Ins2(+/-) mice remained lighter than Ins1(-/-):Ins2(+/+) littermates despite reaching an equivalent degree of hyperinsulinaemia on HFD by 52 weeks. CONCLUSIONS/INTERPRETATION These data demonstrate that attenuation of hyperinsulinaemia in young, growing female mice provides a long-lasting protection against obesity. This protection persists despite a late-onset emergence of hyperinsulinaemia in HFD-fed Ins1(-/-):Ins2(+/-) mice. Given the evolutionary conserved roles of insulin, it is possible that suppressing hyperinsulinaemia early in life may have far-reaching consequences on obesity in full-grown adult humans.
Collapse
Affiliation(s)
- Nicole M Templeman
- Diabetes Research Group, Life Sciences Institute, Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3
| | - Susanne M Clee
- Diabetes Research Group, Life Sciences Institute, Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3
| | - James D Johnson
- Diabetes Research Group, Life Sciences Institute, Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3.
| |
Collapse
|
48
|
Yin F, Sancheti H, Liu Z, Cadenas E. Mitochondrial function in ageing: coordination with signalling and transcriptional pathways. J Physiol 2015; 594:2025-42. [PMID: 26293414 DOI: 10.1113/jp270541] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 08/13/2015] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction entailing decreased energy-transducing capacity and perturbed redox homeostasis is an early and sometimes initiating event in ageing and age-related disorders involving tissues with high metabolic rate such as brain, liver and heart. In the central nervous system (CNS), recent findings from our and other groups suggest that the mitochondrion-centred hypometabolism is a key feature of ageing brains and Alzheimer's disease. This hypometabolic state is manifested by lowered neuronal glucose uptake, metabolic shift in the astrocytes, and alternations in mitochondrial tricarboxylic acid cycle function. Similarly, in liver and adipose tissue, mitochondrial capacity around glucose and fatty acid metabolism and thermogenesis is found to decline with age and is implicated in age-related metabolic disorders such as obesity and type 2 diabetes mellitus. These mitochondrion-related disorders in peripheral tissues can impact on brain functions through metabolic, hormonal and inflammatory signals. At the cellular level, studies in CNS and non-CNS tissues support the notion that instead of being viewed as autonomous organelles, mitochondria are part of a dynamic network with close interactions with other cellular components through energy- or redox-sensitive cytosolic kinase signalling and transcriptional pathways. Hence, it would be critical to further understand the molecular mechanisms involved in the communication between mitochondria and the rest of the cell. Therapeutic strategies that effectively preserves or improve mitochondrial function by targeting key component of these signalling cascades could represent a novel direction for numerous mitochondrion-implicated, age-related disorders.
Collapse
Affiliation(s)
- Fei Yin
- Pharmacology & Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA
| | - Harsh Sancheti
- Pharmacology & Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA
| | - Zhigang Liu
- Pharmacology & Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA
| | - Enrique Cadenas
- Pharmacology & Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA
| |
Collapse
|
49
|
Angione C, Costanza J, Carapezza G, Lió P, Nicosia G. Multi-Target Analysis and Design of Mitochondrial Metabolism. PLoS One 2015; 10:e0133825. [PMID: 26376088 PMCID: PMC4574446 DOI: 10.1371/journal.pone.0133825] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 07/02/2015] [Indexed: 12/30/2022] Open
Abstract
Analyzing and optimizing biological models is often identified as a research priority in biomedical engineering. An important feature of a model should be the ability to find the best condition in which an organism has to be grown in order to reach specific optimal output values chosen by the researcher. In this work, we take into account a mitochondrial model analyzed with flux-balance analysis. The optimal design and assessment of these models is achieved through single- and/or multi-objective optimization techniques driven by epsilon-dominance and identifiability analysis. Our optimization algorithm searches for the values of the flux rates that optimize multiple cellular functions simultaneously. The optimization of the fluxes of the metabolic network includes not only input fluxes, but also internal fluxes. A faster convergence process with robust candidate solutions is permitted by a relaxed Pareto dominance, regulating the granularity of the approximation of the desired Pareto front. We find that the maximum ATP production is linked to a total consumption of NADH, and reaching the maximum amount of NADH leads to an increasing request of NADH from the external environment. Furthermore, the identifiability analysis characterizes the type and the stage of three monogenic diseases. Finally, we propose a new methodology to extend any constraint-based model using protein abundances.
Collapse
Affiliation(s)
- Claudio Angione
- Computer Laboratory-University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| | - Jole Costanza
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia, Milan, Italy
| | - Giovanni Carapezza
- Department of Mathematics and Computer Science-University of Catania, Catania, Italy
| | - Pietro Lió
- Computer Laboratory-University of Cambridge, Cambridge, United Kingdom
| | - Giuseppe Nicosia
- Department of Mathematics and Computer Science-University of Catania, Catania, Italy
| |
Collapse
|
50
|
Fischer B, Schöttl T, Schempp C, Fromme T, Hauner H, Klingenspor M, Skurk T. Inverse relationship between body mass index and mitochondrial oxidative phosphorylation capacity in human subcutaneous adipocytes. Am J Physiol Endocrinol Metab 2015; 309:E380-7. [PMID: 26081284 DOI: 10.1152/ajpendo.00524.2014] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 06/15/2015] [Indexed: 01/14/2023]
Abstract
Obesity is characterized by a substantial increase in adipose tissue that may contribute to energy balance. Recently, obesity was suggested to be associated with impaired mitochondrial function in adipocytes. In this study, we investigated the following: 1) the respiratory capacities of mitochondria isolated from mature adipocytes of female subjects whose body mass index (BMI) values were distributed over a wide range and 2) the amounts of electron transport chain complexes in these mitochondria. Fat cells were isolated from adipose tissue specimens by collagenase digestion. Mitochondria were isolated from these fat cells, and their respiratory capacity was determined using a Clark-type electrode. Fat cells were also sorted on the basis of their size into large and small fractions to assess their respiration. Western blot analyses were performed to quantify respiratory chain complex components. We also examined mitochondrial activity development during differentiation using human Simpson-Golabi-Behmel syndrome cells. Our results showed that mitochondrial respiratory capacities in adipocytes were inversely associated with BMI values but were independent of cell size. Western blot analyses revealed significantly fewer complex I and IV components in adipose tissues from obese compared with nonobese women. These results suggest that differences at the level of respiratory chain complexes might be responsible for the deterioration of respiratory capacity in obese individuals. In particular, electron transport at the level of complexes I and IV seems to be most affected.
Collapse
Affiliation(s)
- Britta Fischer
- Technische Universität München, ZIEL Institute for Food and Health, Nutritional Medicine, Freising-Weihenstephan, Germany
| | - Theresa Schöttl
- Technische Universität München, Molecular Nutritional Medicine, Freising-Weihenstephan, Germany
| | - Christina Schempp
- Technische Universität München, ZIEL Institute for Food and Health, Nutritional Medicine, Freising-Weihenstephan, Germany
| | - Tobias Fromme
- Technische Universität München, Molecular Nutritional Medicine, Freising-Weihenstephan, Germany; Technische Universität München, ZIEL, Molecular Nutritional Medicine, Freising-Weihenstephan, Germany
| | - Hans Hauner
- Technische Universität München, ZIEL Institute for Food and Health, Nutritional Medicine, Freising-Weihenstephan, Germany; Institute of Nutritional Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany; and
| | - Martin Klingenspor
- Technische Universität München, Molecular Nutritional Medicine, Freising-Weihenstephan, Germany; Technische Universität München, ZIEL, Molecular Nutritional Medicine, Freising-Weihenstephan, Germany
| | - Thomas Skurk
- Technische Universität München, ZIEL Institute for Food and Health, Nutritional Medicine, Freising-Weihenstephan, Germany; Institute of Nutritional Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany; and
| |
Collapse
|