1
|
Harland AJ, Perks CM. IGFBP-2 and IGF-II: Key Components of the Neural Stem Cell Niche? Implications for Glioblastoma Pathogenesis. Int J Mol Sci 2025; 26:4749. [PMID: 40429889 PMCID: PMC12111820 DOI: 10.3390/ijms26104749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Glioblastoma is a fatal and aggressive cancer with no cure. It is becoming increasingly clear that glioblastoma initiation is a result of adult neural stem cell (NSC) transformation-most likely those within the subventricular zone (SVZ). Indeed, transcriptomic analysis indicates that glioblastomas are reminiscent of a neurodevelopmental hierarchy, in which neural stem and progenitor markers are widely expressed by tumour stem-like cells. However, NSC fates and the cues that drive them are poorly understood. Studying the crosstalk within NSC niches may better inform our understanding of glioblastoma initiation and development. Insulin-like growth factor binding protein 2 (IGFBP-2) has a well-established prognostic role in glioblastoma, and cell-based mechanistic studies show the independent activation of downstream oncogenic pathways. However, IGFBP-2 is more commonly recognised as a modulator of insulin-like growth factors (IGFs) for receptor tyrosine kinase signal propagation or attenuation. In the adult human brain, both IGFBP-2 and IGF-II expression are retained in the choroid plexus (ChP) and secreted into the cerebral spinal fluid (CSF). Moreover, secretion by closely associated cells and NSCs themselves position IGFBP-2 and IGF-II as interesting factors within the NSC niche. In this review, we will highlight the experimental findings that show IGFBP-2 and IGF-II influence NSC behaviour. Moreover, we will link this to glioblastoma biology and demonstrate the requirement for further analysis of these factors in glioma stem cells (GSCs).
Collapse
Affiliation(s)
| | - Claire M. Perks
- Cancer Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK;
| |
Collapse
|
2
|
Ji F, Lee HS, Lee H, Kim JH. The impact of frailty syndrome on skeletal muscle histology: preventive effects of exercise. FEBS Open Bio 2025. [PMID: 40325953 DOI: 10.1002/2211-5463.70049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/17/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
Frailty syndrome, a condition marked by increased vulnerability due to age-related physiological decline, exerts a profound impact on skeletal muscle structure and function. Despite its widespread prevalence, the underlying mechanisms contributing to frailty-associated muscle deterioration remain poorly elucidated. This study utilized histological and biochemical analyses in a murine model to investigate the effects of frailty syndrome on skeletal muscle. Mice were classified based on age and condition, including a subset subjected to an exercise intervention. Parameters evaluated included body weight, lean mass ratio, myofiber size and number, extracellular matrix (ECM) content, and myosin heavy chain isoform expression. Frailty syndrome led to increased body weight and ECM content, coupled with reductions in myofiber size and number, reflecting substantial structural and functional impairments in skeletal muscle. Exercise interventions effectively countered these deleterious changes, preserving myofiber morphology and reducing ECM expansion, thereby demonstrating the protective role of exercise in mitigating frailty-induced muscle deterioration. The study highlights the severe impact of frailty syndrome on skeletal muscle structure and integrity. Importantly, it underscores the potential of regular exercise as an effective therapeutic approach to prevent or reverse muscle deterioration associated with frailty, offering critical insights into managing age-related muscular degeneration.
Collapse
Affiliation(s)
- Fujue Ji
- Department of Physical Education, College of Performing Arts and Sport, Hanyang University, Seoul, Korea
- BK21 FOUR Human-Tech Convergence Program, Hanyang University, Seoul, Korea
| | - Hae Sung Lee
- Department of Physical Education, College of Education, Wonkwang University, Iksan, Korea
| | - Haesung Lee
- Department of Physical Education, College of Performing Arts and Sport, Hanyang University, Seoul, Korea
- BK21 FOUR Human-Tech Convergence Program, Hanyang University, Seoul, Korea
| | - Jong-Hee Kim
- Department of Physical Education, College of Performing Arts and Sport, Hanyang University, Seoul, Korea
- BK21 FOUR Human-Tech Convergence Program, Hanyang University, Seoul, Korea
| |
Collapse
|
3
|
Zhuo Y, Xu Y, Qu X, Li Q, Sun M, Gao X, Yuan F, Cao M, Pan B. Research on Peripheral Nerve Aging and Degeneration: Cellular Changes and Mechanism Exploration From the Perspective of Single-Cell Sequencing. Eur J Neurosci 2025; 61:e70129. [PMID: 40317786 DOI: 10.1111/ejn.70129] [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: 01/01/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
As age increases, there are structural and functional alterations in the peripheral nervous system (PNS), significantly affecting movement, sensation and autonomic function. Understanding the characteristics and mechanisms of PNS aging is crucial for preventing and treating related diseases. This study employed single-cell sequencing technology to analyse the dorsal root ganglia (DRG) and sciatic nerve (SN) of aging rats, in comparison with adult rats. The research investigated the mechanisms underlying PNS aging and degeneration, revealing the transcriptional profiles of various cell types. Significant differences were observed in the proportion of Schwann cells between the DRG and SN of adult and aged rats. The Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) revealed that pathways related to neurodegeneration were upregulated in Schwann cells. Additionally, lipid metabolism pathways were upregulated in the SN of aged rats, suggesting that certain lipid signalling molecules may influence cell proliferation. Through further re-clustering of myelinating Schwann cells, six distinct subtypes were identified. The anti-aging protein protocadherin 9 (PCDH9) was preliminarily screened and found to be significantly downregulated with age. In vitro experiments confirmed that PCDH9 expression is associated with Schwann cell proliferation and differentiation. By using gene expression analysis and cell type across several age groups, this study offers important insights into the mechanisms of PNS aging and degeneration.
Collapse
Affiliation(s)
- Yuyang Zhuo
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yingcai Xu
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xinzhe Qu
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qupeng Li
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Maji Sun
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiao Gao
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Feng Yuan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Menghan Cao
- Center of Clinical Oncology, The Afffliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Bin Pan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
4
|
Palmer RD. The protein paradox, carnivore diet & hypertrophy versus longevity.: Short term nutrition and hypertrophy versus longevity. Nutr Health 2025:2601060251314575. [PMID: 40094942 DOI: 10.1177/02601060251314575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Meat consumption has been a common food selection for humans for millennia. Meat is rich in amino acids, delivers vast amounts of nutrients and assists in short term health and hypertrophy. However, meat consumption can induce the activation of mTOR and IGF-1, accelerated aging, vascular constriction, atherosclerosis, heart disease, increased risk of diabetes, systemic inflammatory effects, cancers (including colorectal and prostate cancers), advanced glycation end products, impaired immune function / increased susceptibility to infection via downstream advanced glycation end product accumulation, polycyclic aromatic hydrocarbon ingestion, increased homocysteine levels among many other pathophysiologies. Research papers showing health benefits of meat consumption versus other papers showing the detriment of meat have led to confusion as many cohorts such as bodybuilding, health and wellness groups, carnivore diet practitioners, online social media longevity groups and more are interested in data that exists across the peer reviewed literature, however, few papers offer a super wide view where meat consumption benefits and pitfalls are taken into account.BackgroundThe need for such a systematic review is high as health enthusiasts incorrectly often quote single data points from papers showing a single benefit from consuming meat. This often leads to a higher consumption of meat. However, not all meat consumption is the same, and not all meat delivers the same benefits or detriments. Therefore, a systematic review of current literature has been performed to extrapolate the data into whether those interested in hypertrophy, short term nutrition and energy, and longevity should consume meat. Aim: The aim of this research is to dispel myths about meat consumption, such as that meat has a one size fits all benefit to all those that consume it regardless of genetics, or that consuming meat-based protein is the same across all meats.MethodsA deep analysis of almost one hundred peer reviewed papers and surveys spanning decades of cohorts having a meat-based diet compared to those consuming a plant based diet has been performed. Further analysis on specific side effects and disease has also been performed.ResultsThe results of our systematic review show clearly that meat is great for hypertrophy, short term nutrition, short term energy requirements, but a very poor choice when it comes to healthy aging and longevity.ConclusionAnimal protein is great for building muscle, short term energy, maintaining high levels of nutrients, but a carnivore diet holds too many adverse long term side effects to be considered a staple for a longevity-based diet. The evidence is very strong, that subjects interested in longevity and aging should shift their protein intake away from red and processed meats, and either toward white meats or plant-based sources if longevity is the goal.
Collapse
|
5
|
Lyon A, Agius T, Macarthur MR, Kiesworo K, Stavart L, Allagnat F, Mitchell SJ, Riella LV, Uygun K, Yeh H, Déglise S, Golshayan D, Longchamp A. Dietary or pharmacological inhibition of insulin-like growth factor-1 protects from renal ischemia-reperfusion injury in mice. iScience 2024; 27:111256. [PMID: 39759002 PMCID: PMC11700642 DOI: 10.1016/j.isci.2024.111256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/29/2024] [Accepted: 10/23/2024] [Indexed: 01/07/2025] Open
Abstract
One-week protein restriction (PR) limits ischemia-reperfusion (IR) damages and improves metabolic fitness. Similarly, longer-term calory restriction results in increased lifespan, partly via reduced insulin-like growth factor (IGF)-1. However, the influence of short-term PR on IGF-1 and its impact on IR are unknown. PR was achieved in mice via one-week carbohydrate loading and/or through a low-protein diet. PR decreased IGF-1 circulating levels as well as renal and hepatic expression. Upon renal IR, serum IGF-1 positively correlated with renal dysfunction and tissular damages, independently of sex and age. Exogenous IGF-1 administration abrogated PR benefits during IR, while IGF-1 receptor inhibition with linsitinib was protective. IGF-1 was associated with a reduction in forkhead box O (FoxO), and AMP-activated protein kinase (AMPK) signaling pathways previously demonstrated to improve IR resilience in various organs. These data support dietary or pharmacological reduction of IGF-1 signaling to mitigate IR injury prior to solid organ transplantation and beyond.
Collapse
Affiliation(s)
- Arnaud Lyon
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
- Transplantation Center and Transplantation Immunopathology Laboratory, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas Agius
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael R. Macarthur
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Kevin Kiesworo
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Louis Stavart
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
- Transplantation Center and Transplantation Immunopathology Laboratory, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | | | - Leonardo V. Riella
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Korkut Uygun
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi Yeh
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sebastien Déglise
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Déla Golshayan
- Transplantation Center and Transplantation Immunopathology Laboratory, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Ji F, Park J, Rheem H, Kim JH. Overlapping and Distinct Physical and Biological Phenotypes in Pure Frailty and Obese Frailty. Biosci Rep 2024; 44:BSR20240784. [PMID: 39382189 PMCID: PMC11554920 DOI: 10.1042/bsr20240784] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/18/2024] [Accepted: 10/03/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Pure frailty and obese frailty are common types of frailty syndrome. However, the overlapping and distinct characteristics between pure frailty and obese frailty remain unclear. This study aims to reveal the overlapping/distinct physical and biological phenotypes of pure frailty and obese frailty, providing theoretical support for their prevention, diagnosis, and treatment. METHOD Mice were fed either a normal or high-fat diet and assessed at 20 months of age. They were assigned to one of the four groups: control, obesity, pure frailty, and obese frailty. Grip strength, walking speed, physical activity, endurance, and body weight were measured to determine pure frailty and obese frailty. Physical and biological phenotypes were assessed. RESULTS Distinct physical phenotypes were observed between pure frailty and obese frailty in terms of body weight, lean mass, fat mass, fat mass in tissue, grip strength, endurance, and physical activity, while walking speed overlapped. In biological phenotypes, levels of Smad2/3, FoxO3a, P62, LAMP-2, and cathepsin L expression were distinct, while AKT, p-AKT, mTOR, p-mTOR, p-Smad2/3, p-FoxO3a, Beclin-1, ATG7, and LC3 overlapped. CONCLUSION Distinct physical phenotypes observed in obese frailty are primarily attributable to the effect of obesity, with further impairment of muscle function resulting from the combined effects of frailty syndromes and obesity. Pure frailty and obese frailty share overlapping biological phenotypes, particularly in the regulation of muscle protein synthesis. Moreover, the interaction between obesity and frailty syndromes gives rise to both overlapping and distinct biological phenotypes, especially in the regulation of specific degradation signaling proteins.
Collapse
Affiliation(s)
- Fujue Ji
- Major in Sport Science, Division of Sport Industry and Science, College of Performing Arts and Sport, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
- BK21 FOUR Human-Tech Convergence Program, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Ji Hyun Park
- Major in Sport Science, Division of Sport Industry and Science, College of Performing Arts and Sport, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Hyeonseung Rheem
- Major in Sport Science, Division of Sport Industry and Science, College of Performing Arts and Sport, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
- BK21 FOUR Human-Tech Convergence Program, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Jong-Hee Kim
- Major in Sport Science, Division of Sport Industry and Science, College of Performing Arts and Sport, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
- BK21 FOUR Human-Tech Convergence Program, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| |
Collapse
|
7
|
Forni MF, Pizzurro GA, Krause W, Alexander AF, Bridges K, Xu Y, Justynski O, Gabry A, Camara NOS, Miller-Jensen K, Horsley V. Multiomics reveals age-dependent metabolic reprogramming of macrophages by wound bed niche secreted signals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.621159. [PMID: 39553941 PMCID: PMC11565841 DOI: 10.1101/2024.10.30.621159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The cellular metabolism of macrophages depends on tissue niches and can control macrophage inflammatory or resolving phenotypes. Yet, the identity of signals within tissue niches that control macrophage metabolism is not well understood. Here, using single-cell RNA sequencing of macrophages in early mouse wounds, we find that, rather than gene expression of canonical inflammatory or resolving polarization markers, metabolic gene expression defines distinct populations of early wound macrophages. Single-cell secretomics and transcriptomics identify inflammatory and resolving cytokines expressed by early wound macrophages, and we show that these signals drive metabolic inputs and mitochondrial metabolism in an age-dependent manner. We show that aging alters the metabolome of early wound macrophages and rewires their metabolism from mitochondria to glycolysis. We further show that macrophage-derived Chi3l3 and IGF-1 can induce metabolic inputs and mitochondrial mass/metabolism in aged and bone marrow-derived macrophages. Together, these findings reveal that macrophage-derived signals drive the mitochondrial metabolism of macrophages within early wounds in an age-dependent manner and have implications for inflammatory diseases, chronic injuries, and age-related inflammatory diseases. In Brief This study reveals that macrophage subsets in early inflammatory stages of skin wound healing are defined by their metabolic profiles rather than polarization phenotype. Using single-cell secretomics, we establish key macrophage cytokines that comprise the in vivo wound niche and drive mitochondrial-based metabolism. Aging significantly alters macrophage heterogeneity and increases glycolytic metabolism, which can be restored to OxPHOS-based metabolism with young niche cytokines. These findings highlight the importance of the tissue niche in driving macrophage phenotypes, with implications for aging-related impairments in wound healing. Highlights Single cell transcriptional analysis reveals that reveals that metabolic gene expression identifies distinct macrophage populations in early skin wounds.Single-cell secretomic data show that young macrophages contribute to the wound bed niche by secreting molecules such as IGF-1 and Chi3l3.Old wound macrophages display altered metabolomics, elevated glycolytic metabolism and glucose uptake, and reduced lipid uptake and mitochondrial mass/metabolism.Chi3l3 but not IGF-1 secretion is altered in macrophages in an age dependent manner.Chi3l3 can restore mitochondrial mass/metabolism in aged macrophages.
Collapse
|
8
|
Sadanandan J, Sathyanesan M, Newton SS. Aging alters the expression of trophic factors and tight junction proteins in the mouse choroid plexus. Fluids Barriers CNS 2024; 21:77. [PMID: 39334352 PMCID: PMC11438291 DOI: 10.1186/s12987-024-00574-0] [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: 03/18/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The choroid plexus (CP) is an understudied tissue in the central nervous system and is primarily implicated in cerebrospinal fluid (CSF) production. CP also produces numerous neurotrophic factors (NTF) which circulate to different brain regions. Regulation of NTFs in the CP during natural aging is largely unknown. Here, we investigated the age and gender-specific transcription of NTFs along with the changes in the tight junctional proteins (TJPs) and the water channel protein Aquaporin (AQP1). METHODS Male and female mice were used for our study. Age-related transcriptional changes were analyzed using quantitative PCR at three different time points: mature adult, middle-aged, and aged. Transcriptional changes during aging were further confirmed with digital droplet PCR. Additionally, we used immunohistochemical analysis (IHC) for the evaluation of in vivo protein expression. We further investigated the cellular phenotype of these NTFS, TJP, and water channel proteins in the mouse CP by co-labeling them with the classical vascular marker, Isolectin B4, and epithelial cell marker, Plectin. RESULTS Aging significantly altered NTF gene expression in the CP. Brain-derived neurotrophic factor (BDNF), Midkine (MDK), VGF, Insulin-like growth factor (IGF1), IGF2, Klotho (KL), Erythropoietin (EPO), and its receptor (EPOR) were reduced in the aged CP of males and females. Vascular endothelial growth factor (VEGF) transcription was gender-specific; in males, gene expression was unchanged in the aged CP, while females showed an age-dependent reduction. Age-dependent changes in VEGF localization were evident, from vasculature to epithelial cells. IGF2 and klotho localized in the basolateral membrane of the CP and showed an age-dependent reduction in epithelial cells. Water channel protein AQP1 localized in the tip of epithelial cells and showed an age-related reduction in mRNA and protein levels. TJP's JAM, CLAUDIN1, CLAUDIN2 and CLAUDIN5 were reduced in aged mice. CONCLUSIONS Our study highlights transcriptional level changes in the CP during aging. The age-related transcriptional changes exhibit similarities as well as gene-specific differences in the CP of males and females. Altered transcription of the water channel protein AQP1 and TJPs could be involved in reduced CSF production during aging. Importantly, reduction in the neurotrophic factors and longevity factor Klotho can play a role in regulating brain aging.
Collapse
Affiliation(s)
- Jayanarayanan Sadanandan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Monica Sathyanesan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Samuel S Newton
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA.
| |
Collapse
|
9
|
Fertan E, Gendron WH, Wong AA, Hanson GM, Brown RE, Weaver ICG. Noncanonical regulation of imprinted gene Igf2 by amyloid-beta 1-42 in Alzheimer's disease. Sci Rep 2023; 13:2043. [PMID: 36739453 PMCID: PMC9899226 DOI: 10.1038/s41598-023-29248-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Reduced insulin-like growth factor 2 (IGF2) levels in Alzheimer's disease (AD) may be the mechanism relating age-related metabolic disorders to dementia. Since Igf2 is an imprinted gene, we examined age and sex differences in the relationship between amyloid-beta 1-42 (Aβ42) accumulation and epigenetic regulation of the Igf2/H19 gene cluster in cerebrum, liver, and plasma of young and old male and female 5xFAD mice, in frontal cortex of male and female AD and non-AD patients, and in HEK293 cell cultures. We show IGF2 levels, Igf2 expression, histone acetylation, and H19 ICR methylation are lower in females than males. However, elevated Aβ42 levels are associated with Aβ42 binding to Igf2 DMR2, increased DNA and histone methylation, and a reduction in Igf2 expression and IGF2 levels in 5xFAD mice and AD patients, independent of H19 ICR methylation. Cell culture results confirmed the binding of Aβ42 to Igf2 DMR2 increased DNA and histone methylation, and reduced Igf2 expression. These results indicate an age- and sex-related causal relationship among Aβ42 levels, epigenomic state, and Igf2 expression in AD and provide a potential mechanism for Igf2 regulation in normal and pathological conditions, suggesting IGF2 levels may be a useful diagnostic biomarker for Aβ42 targeted AD therapies.
Collapse
Affiliation(s)
- Emre Fertan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - William H Gendron
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Aimée A Wong
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Gabrielle M Hanson
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Brain Repair Centre, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Ian C G Weaver
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada. .,Department of Psychiatry, Dalhousie University, Halifax, NS, B3H 4R2, Canada. .,Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada. .,Brain Repair Centre, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
| |
Collapse
|
10
|
Wong HS, Freeman DA, Zhang Y. Not just a cousin of the naked mole-rat: Damaraland mole-rats offer unique insights into biomedicine. Comp Biochem Physiol B Biochem Mol Biol 2022; 262:110772. [PMID: 35710053 PMCID: PMC10155858 DOI: 10.1016/j.cbpb.2022.110772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 11/26/2022]
Abstract
Evolutionary medicine has been a fast-growing field of biological research in the past decade. One of the strengths of evolutionary medicine is to use non-traditional model organisms which often exhibit unusual characteristics shaped by natural selection. Studying these unusual traits could provide valuable insight to understand biomedical questions, since natural selection likely discovers solutions to those complex biological problems. Because of many unusual traits, the naked mole-rat (NMR) has attracted attention from different research areas such as aging, cancer, and hypoxia- and hypercapnia-related disorders. However, such uniqueness of NMR physiology may sometimes make the translational study to human research difficult. Damaraland mole-rat (DMR) shares multiple characteristics in common with NMR, but shows higher degree of similarity with human in some aspects of their physiology. Research on DMR could therefore offer alternative insights and might bridge the gap between experimental findings from NMR to human biomedical research. In this review, we discuss studies of DMR as an extension of the current set of model organisms to help better understand different aspects of human biology and disease. We hope to encourage researchers to consider studying DMR together with NMR. By studying these two similar but evolutionarily distinct species, we can harvest the power of convergent evolution and avoid the potential biased conclusions based on life-history of a single species.
Collapse
Affiliation(s)
- Hoi-Shan Wong
- Nine Square Therapeutics, South San Francisco, CA 94080, United States of America.
| | - David A Freeman
- Department of Biological Sciences, The University of Memphis, Memphis, TN 38152, United States of America
| | - Yufeng Zhang
- College of Health Sciences, The University of Memphis, Memphis, TN 38152, United States of America.
| |
Collapse
|
11
|
Jones EJ, Chiou S, Atherton PJ, Phillips BE, Piasecki M. Ageing and exercise-induced motor unit remodelling. J Physiol 2022; 600:1839-1849. [PMID: 35278221 PMCID: PMC9314090 DOI: 10.1113/jp281726] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 02/14/2022] [Indexed: 11/08/2022] Open
Abstract
A motor unit (MU) comprises the neuron cell body, its corresponding axon and each of the muscle fibres it innervates. Many studies highlight age-related reductions in the number of MUs, yet the ability of a MU to undergo remodelling and to expand to rescue denervated muscle fibres is also a defining feature of MU plasticity. Remodelling of MUs involves two coordinated processes: (i) axonal sprouting and new branching growth from adjacent surviving neurons, and (ii) the formation of key structures around the neuromuscular junction to resume muscle-nerve communication. These processes rely on neurotrophins and coordinated signalling in muscle-nerve interactions. To date, several neurotrophins have attracted focus in animal models, including brain-derived neurotrophic factor and insulin-like growth factors I and II. Exercise in older age has demonstrated benefits in multiple physiological systems including skeletal muscle, yet evidence suggests this may also extend to peripheral MU remodelling. There is, however, a lack of research in humans due to methodological limitations which are easily surmountable in animal models. To improve mechanistic insight of the effects of exercise on MU remodelling with advancing age, future research should focus on combining methodological approaches to explore the in vivo physiological function of the MU alongside alterations of the localised molecular environment.
Collapse
Affiliation(s)
- Eleanor J. Jones
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC–Versus Arthritis Centre of Excellence for Musculoskeletal Ageing ResearchNottingham NIHR Biomedical Research CentreSchool of MedicineUniversity of NottinghamNottinghamUK
| | - Shin‐Yi Chiou
- School of SportExercise, and Rehabilitation Sciences, MRC‐Versus Arthritis Centre for Musculoskeletal Ageing Research, Centre for Human Brain HealthUniversity of BirminghamBirminghamUK
| | - Philip J. Atherton
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC–Versus Arthritis Centre of Excellence for Musculoskeletal Ageing ResearchNottingham NIHR Biomedical Research CentreSchool of MedicineUniversity of NottinghamNottinghamUK
| | - Bethan E. Phillips
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC–Versus Arthritis Centre of Excellence for Musculoskeletal Ageing ResearchNottingham NIHR Biomedical Research CentreSchool of MedicineUniversity of NottinghamNottinghamUK
| | - Mathew Piasecki
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC–Versus Arthritis Centre of Excellence for Musculoskeletal Ageing ResearchNottingham NIHR Biomedical Research CentreSchool of MedicineUniversity of NottinghamNottinghamUK
| |
Collapse
|
12
|
Importance of IGF-I levels in IVF: potential relevance for growth hormone (GH) supplementation. J Assist Reprod Genet 2022; 39:409-416. [PMID: 35066698 PMCID: PMC8956770 DOI: 10.1007/s10815-021-02379-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/10/2021] [Indexed: 12/28/2022] Open
Abstract
Purpose Growth hormone (GH) supplementation in association with in vitro fertilization (IVF) is worldwide again increasing, even though study outcomes have been discrepant. Since GH acts via insulin-like growth factor-1 (IGF-1), its utilization in IVF would only seem to make sense with low IGF-1. We, therefore, determined whether IGF-I levels affect IVF outcomes. Methods Retrospectively, 302 consecutive first fresh, non-donor IVF cycles were studied, excluding patients on GH supplementation. Patients were divided into 3 subgroups: IGF-1 in lower 25th percentile (group A, < 132 ng/mL, n = 64); 25th–75th percentile (B, 133–202 ng/mL, n = 164), and upper 25th percentile (C, > 202 ng/mL, n = 74). IGF-1 was tested immunochemiluminometric with normal range at 78–270 ng/mL. Because of the study patients’ adverse selection and low pregnancy chances, the main outcome measure for the study was cycle cancellation. Secondary outcomes were oocyte numbers, embryos transferred, pregnancies, and live births. Results Group A was significantly older than B and C (P = 0.019). IGF-1 decreased with increasing age per year by 2.2 ± 0.65 ng/mL (P = 0.0007). FSH was best in group B and worst in A (trend, P = 0.085); AMH was best in B and worst in A (N.S.). Cycle cancellations were lowest in C (11.6%) and highest in A (25.0%; P = 0.042). This significance further improved with age adjustment (P = 0.021). Oocytes, embryo numbers, pregnancies, and live birth rates did not differ, though oocyte numbers trended highest in B. Conclusions Here presented results support the hypothesis that IGF-1 levels affect IVF outcomes. GH treatments, therefore, may be effective only with low IGF-1.
Collapse
|
13
|
Zhu Y, Fang Y, Medina D, Bartke A, Yuan R. Metformin treatment of juvenile mice alters aging-related developmental and metabolic phenotypes. Mech Ageing Dev 2022; 201:111597. [PMID: 34780856 PMCID: PMC8755607 DOI: 10.1016/j.mad.2021.111597] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 01/03/2023]
Abstract
Accumulating evidence suggests that the influence on developmental traits might have long-term effects on aging and health later in life. Metformin is a widely used drug for treating type 2 diabetes and is also used for delaying sexual maturation in girls with precocious puberty. The current report focuses on investigating the effects of metformin on development and metabolic traits. Heterogeneous mice (UM-HET3) were treated with i.p. metformin between the ages of 15 and 56 days. Our results show that body weight and food consumption were increased in both sexes, and sexual maturation was delayed in females. Tail length and circulating insulin-like growth factor 1 (IGF1) levels were significantly increased in both sexes. No significant difference was found in insulin tolerance test, but glucose tolerance was significantly reduced in the males. Circulating adiponectin and insulin levels were altered by metformin treatment in a sex-specific manner. Analysis of quantitative insulin sensitivity check index (QUICKI) suggests that metformin treatment increased insulin sensitivity in female pups, but had opposite effect in male pups. This study revealed that early life metformin treatment alters development and metabolism of mice in both sex-specific and non-specific manners. These effects of metformin may have long-term impacts on aging-related traits.
Collapse
Affiliation(s)
- Yun Zhu
- Deparment of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois School of Medicine, 801 N. Rutledge, P. O. Box 19628, Springfield, Illinois 62794-9628, USA,Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P. O. Box 19628, Springfield, Illinois 62794-9628, USA
| | - Yimin Fang
- Department of Neurology, Southern Illinois University School of Medicine, 801 N. Rutledge, P. O. Box 19628, Springfield, Illinois 62794-9628, USA
| | - David Medina
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P. O. Box 19628, Springfield, Illinois 62794-9628, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P. O. Box 19628, Springfield, Illinois 62794-9628, USA
| | - Rong Yuan
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge, P. O. Box 19628, Springfield, Illinois 62794-9628, USA
| |
Collapse
|
14
|
Szabo Z, Koczka V, Marosvolgyi T, Szabo E, Frank E, Polyak E, Fekete K, Erdelyi A, Verzar Z, Figler M. Possible Biochemical Processes Underlying the Positive Health Effects of Plant-Based Diets-A Narrative Review. Nutrients 2021; 13:2593. [PMID: 34444753 PMCID: PMC8398942 DOI: 10.3390/nu13082593] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/11/2022] Open
Abstract
Plant-based diets are becoming more popular for many reasons, and epidemiological as well as clinical data also suggest that a well-balanced vegan diet can be adopted for the prevention, and in some cases, in the treatment of many diseases. In this narrative review, we provide an overview of the relationships between these diets and various conditions and their potential biochemical background. As whole plant foods are very rich in food-derived antioxidants and other phytochemicals, they have many positive physiological effects on different aspects of health. In the background of the beneficial health effects, several biochemical processes could stand, including the reduced formation of trimethylamine oxide (TMAO) or decreased serum insulin-like growth factor 1 (IGF-1) levels and altered signaling pathways such as mechanistic target of rapamycin (mTOR). In addition, the composition of plant-based diets may play a role in preventing lipotoxicity, avoiding N-glycolylneuraminic acid (Neu5Gc), and reducing foodborne endotoxin intake. In this article, we attempt to draw attention to the growing knowledge about these diets and provide starting points for further research.
Collapse
Affiliation(s)
- Zoltan Szabo
- Institute of Nutritional Sciences and Dietetics, Faculty of Health Sciences, University of Pecs, 7621 Pecs, Hungary; (E.F.); (E.P.); (Z.V.); (M.F.)
| | - Viktor Koczka
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, 7624 Pecs, Hungary; (V.K.); (E.S.)
- Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pecs, 7621 Pecs, Hungary
| | - Tamas Marosvolgyi
- Institute of Bioanalysis, Medical School, University of Pecs, 7624 Pecs, Hungary;
- Szentagothai Research Center, University of Pecs, 7624 Pecs, Hungary
| | - Eva Szabo
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, 7624 Pecs, Hungary; (V.K.); (E.S.)
| | - Eszter Frank
- Institute of Nutritional Sciences and Dietetics, Faculty of Health Sciences, University of Pecs, 7621 Pecs, Hungary; (E.F.); (E.P.); (Z.V.); (M.F.)
| | - Eva Polyak
- Institute of Nutritional Sciences and Dietetics, Faculty of Health Sciences, University of Pecs, 7621 Pecs, Hungary; (E.F.); (E.P.); (Z.V.); (M.F.)
| | - Kata Fekete
- Institute for Translational Medicine, Medical School, University of Pecs, 7624 Pecs, Hungary;
| | - Attila Erdelyi
- Institute of Health Insurance, Faculty of Health Sciences, University of Pecs, 7621 Pecs, Hungary;
| | - Zsofia Verzar
- Institute of Nutritional Sciences and Dietetics, Faculty of Health Sciences, University of Pecs, 7621 Pecs, Hungary; (E.F.); (E.P.); (Z.V.); (M.F.)
| | - Maria Figler
- Institute of Nutritional Sciences and Dietetics, Faculty of Health Sciences, University of Pecs, 7621 Pecs, Hungary; (E.F.); (E.P.); (Z.V.); (M.F.)
- 2nd Department of Internal Medicine and Nephrology Centre, Clinical Centre, University of Pecs, 7624 Pecs, Hungary
| |
Collapse
|
15
|
Gene Expression Profile in Different Age Groups and Its Association with Cognitive Function in Healthy Malay Adults in Malaysia. Cells 2021; 10:cells10071611. [PMID: 34199148 PMCID: PMC8304476 DOI: 10.3390/cells10071611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/13/2021] [Accepted: 06/21/2021] [Indexed: 11/16/2022] Open
Abstract
The mechanism of cognitive aging at the molecular level is complex and not well understood. Growing evidence suggests that cognitive differences might also be caused by ethnicity. Thus, this study aims to determine the gene expression changes associated with age-related cognitive decline among Malay adults in Malaysia. A cross-sectional study was conducted on 160 healthy Malay subjects, aged between 28 and 79, and recruited around Selangor and Klang Valley, Malaysia. Gene expression analysis was performed using a HumanHT-12v4.0 Expression BeadChip microarray kit. The top 20 differentially expressed genes at p < 0.05 and fold change (FC) = 1.2 showed that PAFAH1B3, HIST1H1E, KCNA3, TM7SF2, RGS1, and TGFBRAP1 were regulated with increased age. The gene set analysis suggests that the Malay adult's susceptibility to developing age-related cognitive decline might be due to the changes in gene expression patterns associated with inflammation, signal transduction, and metabolic pathway in the genetic network. It may, perhaps, have important implications for finding a biomarker for cognitive decline and offer molecular targets to achieve successful aging, mainly in the Malay population in Malaysia.
Collapse
|
16
|
McCune S, Promislow D. Healthy, Active Aging for People and Dogs. Front Vet Sci 2021; 8:655191. [PMID: 34164450 PMCID: PMC8215343 DOI: 10.3389/fvets.2021.655191] [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: 01/18/2021] [Accepted: 05/05/2021] [Indexed: 01/08/2023] Open
Abstract
Dogs act as companions who provide us with emotional and physical support. Their shorter lifespans compel us to learn about the challenges and gifts of caring for older individuals. Our companion dogs can be exemplars of healthy or unhealthy aging, and sentinels of environmental factors that might increase or decrease our own healthy lifespan. In recent years, the field of aging has emphasized not just lifespan, but healthspan—the period of healthy, active lifespan. This focus on healthy, active aging is reflected in the World Health Organization's current focus on healthy aging for the next decade and the 2016 Healthy Aging in Action initiative in the US. This paper explores the current research into aging in both people and companion dogs, and in particular, how the relationship between older adults and dogs impacts healthy, active aging for both parties. The human-dog relationship faces many challenges as dogs, and people, age. We discuss potential solutions to these challenges, including suggestions for ways to continue contact with dogs if dog ownership is no longer possible for an older person. Future research directions are outlined in order to encourage the building of a stronger evidence base for the role of dogs in the lives of older adults.
Collapse
Affiliation(s)
- Sandra McCune
- School of Psychology, School of Life Sciences, University of Lincoln, Lincoln, United Kingdom.,Animal Matters Consultancy Ltd., Stamford, United Kingdom
| | - Daniel Promislow
- Department of Lab Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, United States.,Department of Biology, University of Washington, Seattle, WA, United States
| |
Collapse
|
17
|
Tresguerres FGF, Tresguerres IF, Leco I, Clemente C, Rodríguez-Torres R, Torres J, Carballido J, Tresguerres JAF. Growth Hormone As Antiaging Factor in Old Bones. Rejuvenation Res 2021; 24:354-365. [PMID: 33906424 DOI: 10.1089/rej.2020.2369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Aging induces changes in bone. Growth hormone (GH) is reduced by aging, and age-related changes observed in old bones might be due to a decrease in the GH/insulin-like growth factor-I (IGF-I) axis. GH administration on aged individuals is controversial. This study aimed to assess the effect of systemic GH treatment on bone properties, bone metabolism, and bone mineral density (BMD) in long bone of old rats. Aged Wistar rats were treated with GH at a dose of 2 mg/kg/day during 10 weeks. Plasma osteocalcin, IGF-I, and carboxy-terminal telopeptide of type I collagen levels were measured. Cross-sectional bone areas and BMD were measured by morphometric and densitometric analysis, respectively. Femora were analyzed by three point-bending testing. t-Test was used for statistical evaluation. p < 0.05 was considered to be significant. Significantly enhanced bone area, at the expense of the cortical area, was found in treated rats. The densitometric analysis showed 11% higher BMD in the experimental group. Significantly higher bone flexural modulus, stiffness, and ultimate load were observed in the treated rats. Plasma osteocalcin and IGF-I levels were significantly increased in the treated group, while the resorption marker concentration remained unchanged. Within the limitations of this experimental study, systemic GH administration has shown to enhance biomechanical properties, BMD, cortical mass, and plasma IGF-I and osteocalcin in old treated rats, compared to the control group; consequently, GH could be considered as an alternative therapy against age-related changes in the bone.
Collapse
Affiliation(s)
- Francisco G F Tresguerres
- Department of Dental Clinical Specialities, Faculty of Dentistry, Complutense University, Madrid, Spain
| | - Isabel F Tresguerres
- Department of Dental Clinical Specialities, Faculty of Dentistry, Complutense University, Madrid, Spain
| | - Isabel Leco
- Department of Dental Clinical Specialities, Faculty of Dentistry, Complutense University, Madrid, Spain
| | - Celia Clemente
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Alcala, Madrid, Spain
| | - Rosa Rodríguez-Torres
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Alcala, Madrid, Spain
| | - Jesús Torres
- Department of Dental Clinical Specialities, Faculty of Dentistry, Complutense University, Madrid, Spain
| | - Jorge Carballido
- Department of Dental Clinical Specialities, Faculty of Dentistry, Complutense University, Madrid, Spain
| | - Jesús A F Tresguerres
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
18
|
Gasmi A, Chirumbolo S, Peana M, Mujawdiya PK, Dadar M, Menzel A, Bjørklund G. Biomarkers of Senescence during Aging as Possible Warnings to Use Preventive Measures. Curr Med Chem 2021; 28:1471-1488. [PMID: 32942969 DOI: 10.2174/0929867327999200917150652] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 08/09/2020] [Accepted: 08/18/2020] [Indexed: 11/22/2022]
Abstract
Human life expectancy is increasing significantly over time thanks to the improved possibility for people to take care of themselves and the higher availability of food, drugs, hygiene, services, and assistance. The increase in the average age of the population worldwide is, however, becoming a real concern, since aging is associated with the rapid increase in chronic inflammatory pathologies and degenerative diseases, very frequently dependent on senescent phenomena that occur alongside with senescence. Therefore, the search for reliable biomarkers that can diagnose the possible onset or predict the risk of developing a disease associated with aging is a crucial target of current medicine. In this review, we construct a synopsis of the main addressable biomarkers to study the development of aging and the associated ailments.
Collapse
Affiliation(s)
- Amin Gasmi
- Société Francophone de Nutrithérapie et de Nutrigénétique Appliquée, Villeurbanne, France
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Massimiliano Peana
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy
| | | | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Alain Menzel
- Laboratoires Réunis, Junglinster, Luxembourg, Norway
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| |
Collapse
|
19
|
Li N, Zhao S, Zhang Z, Zhu Y, Gliniak CM, Vishvanath L, An YA, Wang MY, Deng Y, Zhu Q, Shan B, Sherwood A, Onodera T, Oz OK, Gordillo R, Gupta RK, Liu M, Horvath TL, Dixit VD, Scherer PE. Adiponectin preserves metabolic fitness during aging. eLife 2021; 10:65108. [PMID: 33904399 PMCID: PMC8099426 DOI: 10.7554/elife.65108] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
Adiponectin is essential for the regulation of tissue substrate utilization and systemic insulin sensitivity. Clinical studies have suggested a positive association of circulating adiponectin with healthspan and lifespan. However, the direct effects of adiponectin on promoting healthspan and lifespan remain unexplored. Here, we are using an adiponectin null mouse and a transgenic adiponectin overexpression model. We directly assessed the effects of circulating adiponectin on the aging process and found that adiponectin null mice display exacerbated age-related glucose and lipid metabolism disorders. Moreover, adiponectin null mice have a significantly shortened lifespan on both chow and high-fat diet. In contrast, a transgenic mouse model with elevated circulating adiponectin levels has a dramatically improved systemic insulin sensitivity, reduced age-related tissue inflammation and fibrosis, and a prolonged healthspan and median lifespan. These results support a role of adiponectin as an essential regulator for healthspan and lifespan.
Collapse
Affiliation(s)
- Na Li
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States.,Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Shangang Zhao
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Zhuzhen Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Yi Zhu
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Christy M Gliniak
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Lavanya Vishvanath
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Yu A An
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - May-Yun Wang
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Yingfeng Deng
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Qingzhang Zhu
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Bo Shan
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Amber Sherwood
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Toshiharu Onodera
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Orhan K Oz
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Tamas L Horvath
- Department of Comparative Medicine and Immunobiology, Yale School of Medicine, New Haven, United States
| | - Vishwa Deep Dixit
- Department of Comparative Medicine and Immunobiology, Yale School of Medicine, New Haven, United States.,Yale Center for Research on Aging, Yale School of Medicine, New Haven, United States
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, United States.,Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
20
|
IGF-1 Upregulates Biglycan and Decorin by Increasing Translation and Reducing ADAMTS5 Expression. Int J Mol Sci 2021; 22:ijms22031403. [PMID: 33573338 PMCID: PMC7866853 DOI: 10.3390/ijms22031403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 12/11/2022] Open
Abstract
Proteoglycan (PG) is a glycosaminoglycan (GAG)-conjugated protein essential for maintaining tissue strength and elasticity. The most abundant skin PGs, biglycan and decorin, have been reported to decrease as skin ages. Insulin-like growth factor-1 (IGF-1) is important in various physiological functions such as cell survival, growth, and apoptosis. It is well known that the serum level of IGF-1 decreases with age. Therefore, we investigated whether and how IGF-1 affects biglycan and decorin. When primary cultured normal human dermal fibroblasts (NHDFs) were treated with IGF-1, protein levels of biglycan and decorin increased, despite no difference in mRNA expression. This increase was not inhibited by transcription blockade using actinomycin D, suggesting that it is mediated by IGF-1-induced enhanced translation. Additionally, both mRNA and protein expression of ADAMTS5, a PG-degrading enzyme, were decreased in IGF-1-treated NHDFs. Knockdown of ADAMTS5 via RNA interference increased protein expression of biglycan and decorin. Moreover, mRNA and protein expression of ADAMTS5 increased in aged human skin tissues compared to young tissue. Overall, IGF-1 increases biglycan and decorin, which is achieved by improving protein translation to increase synthesis and preventing ADAMTS5-mediated degradation. This suggests a new role of IGF-1 as a regulator for biglycan and decorin in skin aging process.
Collapse
|
21
|
Ji J, Jin T, Zhang R, Lou A, Chen Y, Xiang S, Cui C, Yu L, Guan L. The effect of miR-6523a on growth hormone secretion in pituitary cells of Yanbian yellow cattle. CANADIAN JOURNAL OF ANIMAL SCIENCE 2020. [DOI: 10.1139/cjas-2019-0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Yanbian yellow cattle breeding is limited by its slow growth. We previously found that the miRNA miR-6523a is differentially expressed between Yanbian yellow cattle and Han Yan cattle, which differ in growth characteristics. In this study, we evaluated the effects of miR-6523a on growth hormone (GH) secretion in pituitary cells of Yanbian yellow cattle. Bioinformatics analyses using TargetScan and RNAhybrid, as well as dual luciferase reporter assays, showed that miR-6523a targets the 3′ untranslated region of somatostatin receptor 5 (SSTR5). We further found that the mRNA and protein expression levels of GH in pituitary cells were significantly higher in cells treated with miR-6523a mimic than in the control group (P = 0.0082 and P = 0.0069). The GH mRNA and protein expression levels were lower in cells treated with miR-6523a inhibitor than in the control group, but the difference was not significant (P = 0.064 and P = 0.089). SSTR5 mRNA and protein levels were inhibited by miR-6523a mimic compared with the control group (P = 0.0024 and P = 0.0028) and were elevated slightly by miR-6523a inhibitor (P = 0.093 and P = 0.091). These results prove that miR-6523a regulates GH secretion in pituitary cells by SSTR5. More broadly, these findings provide a basis for studies of the roles of miRNAs in animal growth and development.
Collapse
Affiliation(s)
- Jiuxiu Ji
- College of Agriculture and Forestry Science, Linyi University, Shuangling Road, Linyi 276005, People’s Republic of China
| | - Taihua Jin
- College of Agriculture and Forestry Science, Linyi University, Shuangling Road, Linyi 276005, People’s Republic of China
| | - Rui Zhang
- Agriculture College, Yanbian University, Yanji 133000, People’s Republic of China
| | - Angang Lou
- Agriculture College, Yanbian University, Yanji 133000, People’s Republic of China
| | - Yingying Chen
- Agriculture College, Yanbian University, Yanji 133000, People’s Republic of China
| | - Siyu Xiang
- Agriculture College, Yanbian University, Yanji 133000, People’s Republic of China
| | - Changyan Cui
- Agriculture College, Yanbian University, Yanji 133000, People’s Republic of China
| | - Longzheng Yu
- Agriculture College, Yanbian University, Yanji 133000, People’s Republic of China
| | - Lizeng Guan
- College of Agriculture and Forestry Science, Linyi University, Shuangling Road, Linyi 276005, People’s Republic of China
| |
Collapse
|
22
|
Short-term fasting differentially regulates PI3K/AkT/mTOR and ERK signalling in the rat hypothalamus. Mech Ageing Dev 2020; 192:111358. [PMID: 32961167 DOI: 10.1016/j.mad.2020.111358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/12/2020] [Accepted: 09/13/2020] [Indexed: 11/20/2022]
Abstract
It is known that insulin secreted by pancreatic β-cells enters the brain by crossing the blood-brain barrier. However, it was demonstrated that insulin expression occurs in various brain regions as well. Albeit the list of insulin actions in the brain is long and it includes control of energy homeostasis, neuronal survival, maintenance of synaptic plasticity and cognition, not much is known about the adaptive significance of insulin synthesis in brain. We previously reported that short-term fasting promotes insulin expression and subsequent activation of insulin receptor in the rat periventricular nucleus. In order to uncover a physiological importance of the fasting-induced insulin expression in hypothalamus, we analyzed the effect of short-term food deprivation on the expression of several participants of PI3K/AKT/mTOR and Ras/MAPK signaling pathways that are typically activated by this hormone. We found that the hypothalamic content of total and activated IRS1, IRS2, PI3K, and mTOR remained unchanged, but phosphorylated AKT1/2/3 was decreased. The levels of activated ERK1/2 were increased after six-hour fasting. Moreover, activated ERK1/2 was co-expressed with activated insulin receptor in the nucleus arcuatus. Our previously published and current findings suggest that the ERK activation in hypothalamus was at least partially initiated by the centrally produced insulin.
Collapse
|
23
|
Ji J, Jin T, Lou A, Zhang R, Chen Y, Xiang S, Cui C, Yu L, Guan L. The effect of miR-10b on growth hormone in pituitary cells of Yanbian yellow cattle by somatostatin receptor 2. Anim Sci J 2020; 91:e13420. [PMID: 32618083 DOI: 10.1111/asj.13420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022]
Abstract
This study aimed to evaluate the effect of miR-10b on growth hormone (GH) in pituitary cells of Yanbian yellow cattle. According to analysis of GH and somatostatin receptor 2 (SSTR2) mRNA and protein expression levels, we found that miR-10b targeted 3'UTR of SSTR2. Compared with the negative control (NC) group, GH mRNA transcription and protein expression in pituitary cells of Yanbian yellow cattle were significantly increased by adding miR-10b mimics (p < .01), while these were significantly decreased by adding miR-10b inhibitor (p < .05); compared with the NC group, SSTR2 mRNA transcription and protein expression were significantly inhibited by the addition of miR-10b mimics (p < .01), while these were significantly increased by the addition of miR-10b inhibitor compared with the iNC group (p < .05). This study suggested that miR-10b could regulate GH level by regulating SSTR2 gene expression in pituitary cells of Yanbian yellow cattle.
Collapse
Affiliation(s)
- Jiuxiu Ji
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Taihua Jin
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Angang Lou
- Agriculture College, Yanbian University, Yanji, China
| | - Rui Zhang
- Agriculture College, Yanbian University, Yanji, China
| | - Yingying Chen
- Agriculture College, Yanbian University, Yanji, China
| | - Siyu Xiang
- Agriculture College, Yanbian University, Yanji, China
| | - Changyan Cui
- Agriculture College, Yanbian University, Yanji, China
| | - Longzheng Yu
- Agriculture College, Yanbian University, Yanji, China
| | - Lizeng Guan
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| |
Collapse
|
24
|
Shen T, Li H, Song Y, Li L, Lin J, Wei G, Ni T. Alternative polyadenylation dependent function of splicing factor SRSF3 contributes to cellular senescence. Aging (Albany NY) 2020; 11:1356-1388. [PMID: 30835716 PMCID: PMC6428108 DOI: 10.18632/aging.101836] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 02/17/2019] [Indexed: 12/18/2022]
Abstract
Down-regulated splicing factor SRSF3 is known to promote cellular senescence, an important biological process in preventing cancer and contributing to individual aging, via its alternative splicing dependent function in human cells. Here we discovered alternative polyadenylation (APA) dependent function of SRSF3 as a novel mechanism explaining SRSF3 downregulation induced cellular senescence. Knockdown of SRSF3 resulted in preference usage of proximal poly(A) sites and thus global shortening of 3′ untranslated regions (3′ UTRs) of mRNAs. SRSF3-depletion also induced senescence-related phenotypes in both human and mouse cells. These 3′ UTR shortened genes were enriched in senescence-associated pathways. Shortened 3′ UTRs tended to produce more proteins than the longer ones. Simulating the effects of 3′ UTR shortening by overexpression of three candidate genes (PTEN, PIAS1 and DNMT3A) all led to senescence-associated phenotypes. Mechanistically, SRSF3 has higher binding density near proximal poly(A) site than distal one in 3′ UTR shortened genes. Further, upregulation of PTEN by either ectopic overexpression or SRSF3-knockdown induction both led to reduced phosphorylation of AKT and ultimately senescence-associated phenotypes. We revealed for the first time that reduced SRSF3 expression could promote cellular senescence through its APA-dependent function, largely extending our mechanistic understanding in splicing factor regulated cellular senescence.
Collapse
Affiliation(s)
- Ting Shen
- State Key Laboratory of Genetic Engineering and Ministry of Education (MOE) Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai 200438, China
| | - Huan Li
- State Key Laboratory of Genetic Engineering and Ministry of Education (MOE) Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai 200438, China
| | - Yifang Song
- State Key Laboratory of Genetic Engineering and Ministry of Education (MOE) Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai 200438, China
| | - Li Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Jinzhong Lin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Gang Wei
- State Key Laboratory of Genetic Engineering and Ministry of Education (MOE) Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai 200438, China
| | - Ting Ni
- State Key Laboratory of Genetic Engineering and Ministry of Education (MOE) Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai 200438, China
| |
Collapse
|
25
|
Pretsch G, Sanadgol N, Smidak R, Lubec J, Korz V, Höger H, Zappe K, Cichna‑Markl M, Lubec G. Doublecortin and IGF-1R protein levels are reduced in spite of unchanged DNA methylation in the hippocampus of aged rats. Amino Acids 2020; 52:543-553. [DOI: 10.1007/s00726-020-02834-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 03/04/2020] [Indexed: 11/24/2022]
|
26
|
Insulin-like growth factor 1 is related to the expression of plumage traits in a passerine species. Behav Ecol Sociobiol 2020. [DOI: 10.1007/s00265-020-2821-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Abstract
Avian plumage colors and ornaments are excellent models to study the endocrine mechanisms linking sexually selected traits and individual parameters of quality and condition. Insulin-like growth factor 1 (IGF-1) is an evolutionarily highly conserved peptide hormone. Its regulatory role in cell proliferation and differentiation and its high sensitivity to the nutritional state of individuals suggest it as an interesting candidate, possibly providing a link between body condition and individual capacity to grow elaborated ornamental features. We investigated whether IGF-1 levels during molting correlate with the expression of multiple ornaments in a sexually dichromatic passerine species, the bearded reedling (Panurus biarmicus). We collected blood samples of males and females shortly before the molting completed and measured the size and colors of ornamental traits. Our results indicate that in males, structural plumage colors, the size of the melanin-based ornament (beard), and tail length are independent traits. IGF-1 levels are associated with the length of the tail and the expression of male structural plumage components (UV coloration), but not the melanin-based ornament. In females, plumage color and tail length were independent traits, which were not related to IGF-1 levels. To the best of our knowledge, this study provides the first evidence that IGF-1 could play a role in the development of secondary sexual characters in a bird species.
Significance statement
IGF-1 is an evolutionarily highly conserved peptide hormone, which recently entered the center stage of research enquiry in evolutionary biology. It is considered as one of the key factors shaping individual life histories, but little is known about its effects on sexually selected traits. We investigated whether IGF-1 levels during molting predict the elaboration of multiple ornamental plumage traits in male and female bearded reedlings (Panurus biarmicus). Our results indicate that higher IGF-1 levels had positive effects on male structural plumage colors and tail feather length. This is the first study, bringing indication for a potential role of IGF-1 in the expression of plumage ornaments in a bird species. Our findings suggest that IGF-1 might serve as an ideal candidate to study the mechanisms linking condition and the capacity to develop sexually selected ornaments.
Collapse
|
27
|
Herrera ML, Basmadjian OM, Falomir‐Lockhart E, Dolcetti FJ, Hereñú CB, Bellini MJ. Sex frailty differences in ageing mice: Neuropathologies and therapeutic projections. Eur J Neurosci 2020; 52:2827-2837. [DOI: 10.1111/ejn.14703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 02/04/2020] [Accepted: 02/09/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Macarena Lorena Herrera
- Departamento de Farmacología Facultad de Ciencias Químicas Instituto de Farmacología Experimental Córdoba (IFEC‐CONICET) Universidad Nacional de Córdoba Córdoba Argentina
- Facultad de Ciencias Médicas Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP‐CONICET) Universidad Nacional de La Plata Buenos Aires Argentina
| | - Osvaldo Martin Basmadjian
- Departamento de Farmacología Facultad de Ciencias Químicas Instituto de Farmacología Experimental Córdoba (IFEC‐CONICET) Universidad Nacional de Córdoba Córdoba Argentina
| | - Eugenia Falomir‐Lockhart
- Facultad de Ciencias Médicas Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP‐CONICET) Universidad Nacional de La Plata Buenos Aires Argentina
| | - Franco Juan‐Cruz Dolcetti
- Facultad de Ciencias Médicas Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP‐CONICET) Universidad Nacional de La Plata Buenos Aires Argentina
| | - Claudia Beatriz Hereñú
- Departamento de Farmacología Facultad de Ciencias Químicas Instituto de Farmacología Experimental Córdoba (IFEC‐CONICET) Universidad Nacional de Córdoba Córdoba Argentina
| | - María José Bellini
- Facultad de Ciencias Médicas Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP‐CONICET) Universidad Nacional de La Plata Buenos Aires Argentina
| |
Collapse
|
28
|
Baumann CW, Kwak D, Thompson LV. Assessing onset, prevalence and survival in mice using a frailty phenotype. Aging (Albany NY) 2019; 10:4042-4053. [PMID: 30562163 PMCID: PMC6326660 DOI: 10.18632/aging.101692] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/28/2018] [Indexed: 11/25/2022]
Abstract
Little is known whether frailty assessments in mice are capable of distinguishing important characteristics of the frailty syndrome. The goals of this study were to identify the onset and the prevalence of frailty across the lifespan and to determine if a frailty phenotype predicts mortality. Body weight, walking speed, strength, endurance and physical activity were assessed in male C57BL/6 mice every three months starting at 14 months of age. Mice that fell in the bottom 20% for walking speed, strength, endurance and physical activity, and in the top 20% for body weight were considered to have a positive frailty marker. The onset of frailty occurred at 17 months, and represented only 3.5% of the mouse cohort. The percentage of frail mice increased with age until basically every mouse was identified as frail. Frail, pre-frail, and non-frail mice had mean survival ages of 27, 29 and 34 months, respectively. In closing, frail mice lack resilience; in that, multiple tissue/organ systems may deteriorate at an accelerated rate and ultimately lead to early mortality when compared to non-frail mice. Identifying the onset and prevalence of frailty, in addition to predicting mortality, has potential to yield information about several aging processes.
Collapse
Affiliation(s)
- Cory W Baumann
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| | - Dongmin Kwak
- Department of Physical Therapy and Athletic Training, Boston University, Boston, MA 02215, USA
| | - LaDora V Thompson
- Department of Physical Therapy and Athletic Training, Boston University, Boston, MA 02215, USA
| |
Collapse
|
29
|
Innate sensitivity to stress facilitates inflammation, alters metabolism and shortens lifespan in a mouse model of social hierarchy. Aging (Albany NY) 2019; 11:9901-9911. [PMID: 31707362 PMCID: PMC6874436 DOI: 10.18632/aging.102440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023]
Abstract
It is known that stress alters homeostasis and may lead to accelerated aging. However, little is known about the contribution of innate susceptibility to stress to the deterioration of physiological functions, acceleration of aging and developing of age-related diseases. By using socially-submissive stress susceptible (Sub) and socially-dominant stress resilient (Dom) selectively bred mouse model we observed a marked reduction in the lifespan of both male and female Sub mice. We found that innate susceptibility to stress correlates with chronic inflammation, development of splenomegaly and a significant increase in the levels of circulating pro-inflammatory cytokines IL-1β and IL-6. Furthermore, Sub mice showed a marked hypoglycemia, reduction of insulin levels, increase in GSK3 activity and elevation of IGF-1 serum levels, as well as low skin surface temperature and body weight. Interestingly, lifelong exposure of Sub mice to chronic mild stress did not further reduce their lifespan, indicating a high level of intrinsic stress. Taken together, our data reveal that social submissiveness coupled with innate stress sensitivity coincides with inflammation, leading to the deterioration of physiological functions and early aging independent of whether an individual is exposed to stress or not.
Collapse
|
30
|
Wang X, Darcy J, Cai C, Jin J, Bartke A, Cao D. Intestinal immunity in hypopituitary dwarf mice: effects of age. Aging (Albany NY) 2019; 10:358-370. [PMID: 29500333 PMCID: PMC5892686 DOI: 10.18632/aging.101393] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 02/23/2018] [Indexed: 12/30/2022]
Abstract
Hypopituitary dwarf mice demonstrate advantages of longevity, but little is known of their colon development and intestinal immunity. Herein we found that Ames dwarf mice have shorter colon and colonic crypts, but larger ratio of mesenteric lymph nodes (MLNs) over body weight than age-matched wild type (WT) mice. In the colonic lamina propria (cLP) of juvenile Ames mice, more inflammatory neutrophils (Ā: 0.15% vs. 0.03% in WT mice) and monocytes (Ā: 7.97% vs. 5.15%) infiltrated, and antigen presenting cells CD11c+ dendritic cells (Ā: 1.39% vs. 0.87%), CD11b+ macrophages (Ā: 3.22% vs. 0.81%) and gamma delta T (γδ T) cells (Ā: 5.56% vs. 1.35%) were increased. In adult Ames dwarf mice, adaptive immune cells, such as IL-17 producing CD4+ T helper (Th17) cells (Ā: 8.3% vs. 4.7%) were augmented. In the MLNs of Ames dwarf mice, the antigen presenting and adaptive immune cells also altered when compared to WT mice, such as a decrease of T-regulatory (Treg) cells in juvenile Ames mice (Ā: 7.7% vs.10.5%), but an increase of Th17 cells (Ā: 0.627% vs.0.093%). Taken together, these data suggest that somatotropic signaling deficiency influences colon development and intestinal immunity.
Collapse
Affiliation(s)
- Xin Wang
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University, School of Medicine, Springfield, IL 62702, USA
| | - Justin Darcy
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University, School of Medicine, Springfield, IL 62702, USA.,Department of Internal Medicine, Southern Illinois University, School of Medicine, Springfield, IL 62702, USA
| | - Chuan Cai
- Division of Stem Cell Regulation and Application, State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan (incubation), Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Junfei Jin
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin 541001, Guangxi, China
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University, School of Medicine, Springfield, IL 62702, USA
| | - Deliang Cao
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University, School of Medicine, Springfield, IL 62702, USA.,Division of Stem Cell Regulation and Application, State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan (incubation), Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| |
Collapse
|
31
|
Farias Quipildor GE, Mao K, Hu Z, Novaj A, Cui MH, Gulinello M, Branch CA, Gubbi S, Patel K, Moellering DR, Tarantini S, Kiss T, Yabluchanskiy A, Ungvari Z, Sonntag WE, Huffman DM. Central IGF-1 protects against features of cognitive and sensorimotor decline with aging in male mice. GeroScience 2019; 41:185-208. [PMID: 31076997 DOI: 10.1007/s11357-019-00065-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/08/2019] [Indexed: 12/22/2022] Open
Abstract
Disruptions in growth hormone/insulin-like growth factor-1 (GH/IGF-1) signaling have been linked to improved longevity in mice and humans. Nevertheless, while IGF-1 levels are associated with increased cancer risk, they have been paradoxically implicated with protection from other age-related conditions, particularly in the brain, suggesting that strategies aimed at selectively increasing central IGF-1 action may have favorable effects on aging. To test this hypothesis, we generated inducible, brain-specific (TRE-IGF-1 × Camk2a-tTA) IGF-1 (bIGF-1) overexpression mice and studied effects on healthspan. Doxycycline was removed from the diet at 12 weeks old to permit post-development brain IGF-1 overexpression, and animals were monitored up to 24 months. Brain IGF-1 levels were increased approximately twofold in bIGF-1 mice, along with greater brain weights, volume, and myelin density (P < 0.05). Age-related changes in rotarod performance, exercise capacity, depressive-like behavior, and hippocampal gliosis were all attenuated specifically in bIGF-1 male mice (P < 0.05). However, chronic brain IGF-1 failed to prevent declines in cognitive function or neurovascular coupling. Therefore, we performed a short-term intranasal (IN) treatment of either IGF-1 or saline in 24-month-old male C57BL/6 mice and found that IN IGF-1 treatment tended to reduce depressive (P = 0.09) and anxiety-like behavior (P = 0.08) and improve motor coordination (P = 0.07) and unlike transgenic mice improved motor learning (P < 0.05) and visuospatial and working memory (P < 0.05). These data highlight important sex differences in how brain IGF-1 action impacts healthspan and suggest that translational approaches that target IGF-1 centrally can restore cognitive function, a possibility that should be explored as a strategy to combat age-related cognitive decline.
Collapse
Affiliation(s)
- Gabriela E Farias Quipildor
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer Bldg, Rm 236, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kai Mao
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer Bldg, Rm 236, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Zunju Hu
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer Bldg, Rm 236, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ardijana Novaj
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer Bldg, Rm 236, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Min-Hui Cui
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Gulinello
- Behavioral Core Facility, Dominick S. Purpura Department of Neuroscience, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY, USA
| | - Craig A Branch
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sriram Gubbi
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Internal Medicine, Jacobi Medical Center, Bronx, NY, USA
| | - Khushbu Patel
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer Bldg, Rm 236, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Douglas R Moellering
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tamas Kiss
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - William E Sonntag
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Derek M Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer Bldg, Rm 236, 1300 Morris Park Avenue, Bronx, NY, 10461, USA. .,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA. .,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
32
|
Ziegler AN, Feng Q, Chidambaram S, Testai JM, Kumari E, Rothbard DE, Constancia M, Sandovici I, Cominski T, Pang K, Gao N, Wood TL, Levison SW. Insulin-like Growth Factor II: An Essential Adult Stem Cell Niche Constituent in Brain and Intestine. Stem Cell Reports 2019; 12:816-830. [PMID: 30905741 PMCID: PMC6450461 DOI: 10.1016/j.stemcr.2019.02.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 02/02/2023] Open
Abstract
Tissue-specific stem cells have unique properties and growth requirements, but a small set of juxtacrine and paracrine signals have been identified that are required across multiple niches. Whereas insulin-like growth factor II (IGF-II) is necessary for prenatal growth, its role in adult stem cell physiology is largely unknown. We show that loss of Igf2 in adult mice resulted in a ∼50% reduction in slowly dividing, label-retaining cells in the two regions of the brain that harbor neural stem cells. Concordantly, induced Igf2 deletion increased newly generated neurons in the olfactory bulb accompanied by hyposmia, and caused impairments in learning and memory and increased anxiety. Induced Igf2 deletion also resulted in rapid loss of stem and progenitor cells in the crypts of Lieberkühn, leading to body-weight loss and lethality and the inability to produce organoids in vitro. These data demonstrate that IGF-II is critical for multiple adult stem cell niches.
Collapse
Affiliation(s)
- Amber N. Ziegler
- Department Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Qiang Feng
- Department of Biological Sciences, Rutgers University-Newark, Newark, NJ 07102, USA
| | - Shravanthi Chidambaram
- Department Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Jaimie M. Testai
- Department Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Ekta Kumari
- Department Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Deborah E. Rothbard
- Department Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Miguel Constancia
- University of Cambridge Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Department of Obstetrics and Gynaecology, Cambridge CB2 0SW, UK,National Institute for Health Research Cambridge Biomedical Research Centre, The University of Cambridge, Cambridge CB2 0SW, UK,Centre for Trophoblast Research, The University of Cambridge, Cambridge CB2 0SW, UK
| | - Ionel Sandovici
- University of Cambridge Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Department of Obstetrics and Gynaecology, Cambridge CB2 0SW, UK,Centre for Trophoblast Research, The University of Cambridge, Cambridge CB2 0SW, UK
| | - Tara Cominski
- Department Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Kevin Pang
- Department Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University-Newark, Newark, NJ 07102, USA
| | - Teresa L. Wood
- Department Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Steven W. Levison
- Department Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA,Corresponding author
| |
Collapse
|
33
|
Weissleder C, Barry G, Fung SJ, Wong MW, Double KL, Webster MJ, Weickert CS. Reduction in IGF1 mRNA in the Human Subependymal Zone During Aging. Aging Dis 2019; 10:197-204. [PMID: 30705779 PMCID: PMC6345338 DOI: 10.14336/ad.2018.0317] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 03/17/2018] [Indexed: 01/09/2023] Open
Abstract
The cell proliferation marker, Ki67 and the immature neuron marker, doublecortin are both expressed in the major human neurogenic niche, the subependymal zone (SEZ), but expression progressively decreases across the adult lifespan (PMID: 27932973). In contrast, transcript levels of several mitogens (transforming growth factor α, epidermal growth factor and fibroblast growth factor 2) do not decline with age in the human SEZ, suggesting that other growth factors may contribute to the reduced neurogenic potential. While insulin like growth factor 1 (IGF1) regulates neurogenesis throughout aging in the mouse brain, the extent to which IGF1 and IGF family members change with age and relate to adult neurogenesis markers in the human SEZ has not yet been determined. We used quantitative polymerase chain reaction to examine gene expression of seven IGF family members [IGF1, IGF1 receptor, insulin receptor and high-affinity IGF binding proteins (IGFBPs) 2, 3, 4 and 5] in the human SEZ across the adult lifespan (n=50, 21-103 years). We found that only IGF1 expression significantly decreased with increasing age. IGFBP2 and IGFBP4 expression positively correlated with Ki67 mRNA. IGF1 expression positively correlated with doublecortin mRNA, whereas IGFBP2 expression negatively correlated with doublecortin mRNA. Our results suggest IGF family members are local regulators of neurogenesis and indicate that the age-related reduction in IGF1 mRNA may limit new neuron production by restricting neuronal differentiation in the human SEZ.
Collapse
Affiliation(s)
- Christin Weissleder
- 1Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia.,2School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Guy Barry
- 3QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Samantha J Fung
- 1Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia.,2School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Matthew W Wong
- 1Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia.,2School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,4School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Kay L Double
- 5Discipline of Biomedical Science and Brain and Mind Centre, Sydney Medical School, University of Sydney, Australia
| | - Maree J Webster
- 6Laboratory of Brain Research, Stanley Medical Research Institute, Maryland, USA
| | - Cynthia Shannon Weickert
- 1Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia.,2School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
34
|
Gubbi S, Quipildor GF, Barzilai N, Huffman DM, Milman S. 40 YEARS of IGF1: IGF1: the Jekyll and Hyde of the aging brain. J Mol Endocrinol 2018; 61:T171-T185. [PMID: 29739805 PMCID: PMC5988994 DOI: 10.1530/jme-18-0093] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 12/30/2022]
Abstract
The insulin-like growth factor 1 (IGF1) signaling pathway has emerged as a major regulator of the aging process, from rodents to humans. However, given the pleiotropic actions of IGF1, its role in the aging brain remains complex and controversial. While IGF1 is clearly essential for normal development of the central nervous system, conflicting evidence has emerged from preclinical and human studies regarding its relationship to cognitive function, as well as cerebrovascular and neurodegenerative disorders. This review delves into the current state of the evidence examining the role of IGF1 in the aging brain, encompassing preclinical and clinical studies. A broad examination of the data indicates that IGF1 may indeed play opposing roles in the aging brain, depending on the underlying pathology and context. Some evidence suggests that in the setting of neurodegenerative diseases that manifest with abnormal protein deposition in the brain, such as Alzheimer's disease, reducing IGF1 signaling may serve a protective role by slowing disease progression and augmenting clearance of pathologic proteins to maintain cellular homeostasis. In contrast, inducing IGF1 deficiency has also been implicated in dysregulated function of cognition and the neurovascular system, suggesting that some IGF1 signaling may be necessary for normal brain function. Furthermore, states of acute neuronal injury, which necessitate growth, repair and survival signals to persevere, typically demonstrate salutary effects of IGF1 in that context. Appreciating the dual, at times opposing 'Dr Jekyll' and 'Mr Hyde' characteristics of IGF1 in the aging brain, will bring us closer to understanding its impact and devising more targeted IGF1-related interventions.
Collapse
Affiliation(s)
- Sriram Gubbi
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Department of Internal MedicineJacobi Medical Center, Bronx, New York, USA
| | - Gabriela Farias Quipildor
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Department of Molecular PharmacologyAlbert Einstein College of Medicine, Bronx, New York, USA
- Division of EndocrinologyDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Nir Barzilai
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Division of EndocrinologyDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Division of GeriatricsDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of GeneticsAlbert Einstein College of Medicine, Bronx, New York, USA
| | - Derek M Huffman
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Department of Molecular PharmacologyAlbert Einstein College of Medicine, Bronx, New York, USA
- Division of EndocrinologyDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sofiya Milman
- Institute for Aging ResearchAlbert Einstein College of Medicine, Bronx, New York, USA
- Division of EndocrinologyDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Division of GeriatricsDepartment of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
35
|
Ogundele OM, Pardo J, Francis J, Goya RG, Lee CC. A Putative Mechanism of Age-Related Synaptic Dysfunction Based on the Impact of IGF-1 Receptor Signaling on Synaptic CaMKIIα Phosphorylation. Front Neuroanat 2018; 12:35. [PMID: 29867375 PMCID: PMC5960681 DOI: 10.3389/fnana.2018.00035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 04/18/2018] [Indexed: 01/13/2023] Open
Abstract
Insulin-like growth factor 1 receptor (IGF-1R) signaling regulates the activity and phosphorylation of downstream kinases linked to inflammation, neurodevelopment, aging and synaptic function. In addition to the control of Ca2+ currents, IGF-1R signaling modulates the activity of calcium-calmodulin-dependent kinase 2 alpha (CaMKIIα) and mitogen activated protein kinase (MAPK/ErK) through multiple signaling pathways. These proteins (CaMKIIα and MAPK) regulate Ca2+ movement and long-term potentiation (LTP). Since IGF-1R controls the synaptic activity of Ca2+, CaMKIIα and MAPK signaling, the possible mechanism through which an age-dependent change in IGF-1R can alter the synaptic expression and phosphorylation of these proteins in aging needs to be investigated. In this study, we evaluated the relationship between an age-dependent change in brain IGF-1R and phosphorylation of CaMKIIα/MAPK. Furthermore, we elucidated possible mechanisms through which dysregulated CaMKIIα/MAPK interaction may be linked to a change in neurotransmitter processing and synaptic function. Male C57BL/6 VGAT-Venus mice at postnatal days 80 (P80), 365 and 730 were used to study age-related neural changes in two brain regions associated with cognitive function: hippocampus and prefrontal cortex (PFC). By means of high throughput confocal imaging and quantitative immunoblotting, we evaluated the distribution and expression of IGF-1, IGF-1R, CaMKIIα, p-CaMKIIα, MAPK and p-MAPK in whole brain lysate, hippocampus and cortex. Furthermore, we compared protein expression patterns and regional changes at P80, P365 and P730. Ultimately, we determined the relative phosphorylation pattern of CaMKIIα and MAPK through quantification of neural p-CaMKIIα and p-MAPK/ErK, and IGF-1R expression for P80, P365 and P730 brain samples. In addition to a change in synaptic function, our results show a decrease in neural IGF-1/IGF-1R expression in whole brain, hippocampus and cortex of aged mice. This was associated with a significant upregulation of phosphorylated neural MAPK (p-MAPK) and decrease in total brain CaMKIIα (i.e., CaMKIIα and p-CaMKIIα) in the aged brain. Taken together, we showed that brain aging is associated with a change in neural IGF-1/IGF-1R expression and may be linked to a change in phosphorylation of synaptic kinases (CaMKIIα and MAPK) that are involved in the modulation of LTP.
Collapse
Affiliation(s)
- Olalekan M. Ogundele
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Joaquin Pardo
- Institute for Biochemical Research of La Plata, School of Medicine, National University of La Plata, La Plata, Argentina
| | - Joseph Francis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Rodolfo G. Goya
- Institute for Biochemical Research of La Plata, School of Medicine, National University of La Plata, La Plata, Argentina
| | - Charles C. Lee
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
36
|
Ng LT, Gruber J, Moore PK. Is there a role of H 2S in mediating health span benefits of caloric restriction? Biochem Pharmacol 2018; 149:91-100. [PMID: 29360438 DOI: 10.1016/j.bcp.2018.01.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/17/2018] [Indexed: 02/07/2023]
Abstract
Caloric restriction (CR) is a dietary regimen that aims to reduce the intake of total calories while maintaining adequate supply of key nutrients so as to avoid malnutrition. CR is one of only a small number of interventions that show promising outcomes on health span and lifespan across different species. There is growing interest in the development of compounds that might replicate CR-related benefits without actually restricting food intake. Hydrogen sulfide (H2S) is produced inside the bodies of many animals, including humans, by evolutionarily conserved H2S synthesizing enzymes. Endogenous H2S is increasingly recognized as an important gaseous signalling molecule involved in diverse cellular and molecular processes. However, the specific role of H2S in diverse biological processes remains to be elucidated and not all its biological effects are beneficial. Nonetheless, recent evidence suggests that the biological functions of H2S intersect with the network of evolutionarily conserved nutrient sensing and stress response pathways that govern organismal responses to CR. Induction of H2S synthesizing enzymes appears to be a conserved and essential feature of the CR response in evolutionarily distant organisms, including nematodes and mice. Here we review the evidence for a role of H2S in CR and lifespan modulation. H2S releasing drugs, capable of controlled delivery of exogenous H2S, are currently in clinical development. These findings suggest such H2S releasing drugs as a promising novel avenue for the development of CR mimetic compounds.
Collapse
Affiliation(s)
- Li Theng Ng
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore; Yale-NUS College, Science Division, Singapore
| | - Jan Gruber
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Yale-NUS College, Science Division, Singapore.
| | - Philip Keith Moore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore
| |
Collapse
|
37
|
Rodriguez-Perez AI, Borrajo A, Diaz-Ruiz C, Garrido-Gil P, Labandeira-Garcia JL. Crosstalk between insulin-like growth factor-1 and angiotensin-II in dopaminergic neurons and glial cells: role in neuroinflammation and aging. Oncotarget 2017; 7:30049-67. [PMID: 27167199 PMCID: PMC5058663 DOI: 10.18632/oncotarget.9174] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 04/19/2016] [Indexed: 01/06/2023] Open
Abstract
The local renin-angiotensin system (RAS) and insulin-like growth factor 1 (IGF-1) have been involved in longevity, neurodegeneration and aging-related dopaminergic degeneration. However, it is not known whether IGF-1 and angiotensin-II (AII) activate each other. In the present study, AII, via type 1 (AT1) receptors, exacerbated neuroinflammation and dopaminergic cell death. AII, via AT1 receptors, also increased the levels of IGF-1 and IGF-1 receptors in microglial cells. IGF-1 inhibited RAS activity in dopaminergic neurons and glial cells, and also inhibited the AII-induced increase in markers of the M1 microglial phenotype. Consistent with this, IGF-1 decreased dopaminergic neuron death induced by the neurotoxin MPP+ both in the presence and in the absence of glia. Intraventricular administration of AII to young rats induced a significant increase in IGF-1 expression in the nigral region. However, aged rats showed decreased levels of IGF-1 relative to young controls, even though RAS activity is known to be enhanced in aged animals. The study findings show that IGF-1 and the local RAS interact to inhibit or activate neuroinflammation (i.e. transition from the M1 to the M2 phenotype), oxidative stress and dopaminergic degeneration. The findings also show that this mechanism is impaired in aged animals.
Collapse
Affiliation(s)
- Ana I Rodriguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana Borrajo
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Carmen Diaz-Ruiz
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
38
|
Shuang T, Fu M, Yang G, Wu L, Wang R. The interaction of IGF-1/IGF-1R and hydrogen sulfide on the proliferation of mouse primary vascular smooth muscle cells. Biochem Pharmacol 2017; 149:143-152. [PMID: 29248598 DOI: 10.1016/j.bcp.2017.12.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 12/12/2017] [Indexed: 11/25/2022]
Abstract
Hydrogen sulfide (H2S) is mostly produced by cystathionine-gamma-lyase (CSE) in vascular system and it inhibits the proliferation of vascular smooth muscle cells (SMCs). Insulin-like growth factor-1 (IGF-1), via its receptor (IGF-1R), exerts multiple physiological and pathophysiological effects on the vasculature, including stimulating SMC proliferation and migration, and inhibiting SMC apoptosis. Since H2S and IGF-1/IGF-1R have opposite effects on SMC proliferation, it becomes imperative to better understand the interaction of these two signaling mechanisms on SMC proliferation. SMCs isolated from small mesenteric arteries of CSE knockout (KO) and wild-type (WT) mice were used in the present study. The effects of IGF-1 and H2S on SMC proliferation were evaluated with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and bromodeoxyuridine (BrdU) assays. Protein expression was determined by western blot, and H2S-induced protein S-sulfhydration was assessed with a modified biotin switch assay. We found that IGF-1 dose-dependently increased the proliferation of both WT-SMCs and KO-SMCs, and this effect was more significant in KO-SMCs. Supplement of sodium hydrosulfide (NaHS) inhibited IGF-1-induced cell proliferation, while this effect was abolished by blocking IGF-1/IGF-1R signaling with picropodophyllin (PPP) or knocking out of the expression of IGF-1R. H2S significantly down-regulates the expression of IGF-1R, stimulates IGF-1R S-sulfhydration, and attenuates the binding of IGF-1 with IGF-1R. This study provides novel insight on the involvement of IGF-1/IGF-1R in H2S-inhibited SMC proliferation and suggests H2S-based innovative treatment strategies for proliferative cardiovascular diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Tian Shuang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Canada; School of Human Kinetics, Laurentian University, Canada; Department of Biology, Laurentian University, Canada; Health Sciences North Research Institute, Sudbury, Ontario, Canada
| | - Ming Fu
- Cardiovascular and Metabolic Research Unit, Laurentian University, Canada; School of Human Kinetics, Laurentian University, Canada; Health Sciences North Research Institute, Sudbury, Ontario, Canada
| | - Guangdong Yang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Canada; Department of Chemistry and Biochemistry, Laurentian University, Ontario, Canada
| | - Lingyun Wu
- Cardiovascular and Metabolic Research Unit, Laurentian University, Canada; School of Human Kinetics, Laurentian University, Canada; Health Sciences North Research Institute, Sudbury, Ontario, Canada
| | - Rui Wang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Canada; Department of Biology, Laurentian University, Canada.
| |
Collapse
|
39
|
Labandeira-Garcia JL, Costa-Besada MA, Labandeira CM, Villar-Cheda B, Rodríguez-Perez AI. Insulin-Like Growth Factor-1 and Neuroinflammation. Front Aging Neurosci 2017; 9:365. [PMID: 29163145 PMCID: PMC5675852 DOI: 10.3389/fnagi.2017.00365] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 10/23/2017] [Indexed: 12/15/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) effects on aging and neurodegeneration is still controversial. However, it is widely admitted that IGF-1 is involved in the neuroinflammatory response. In peripheral tissues, several studies showed that IGF-1 inhibited the expression of inflammatory markers, although other studies concluded that IGF-1 has proinflammatory functions. Furthermore, proinflammatory cytokines such as TNF-α impaired IGF-1 signaling. In the brain, there are controversial results on effects of IGF-1 in neuroinflammation. In addition to direct protective effects on neurons, several studies revealed anti-inflammatory effects of IGF-1 acting on astrocytes and microglia, and that IGF-1 may also inhibit blood brain barrier permeability. Altogether suggests that the aging-related decrease in IGF-1 levels may contribute to the aging-related pro-inflammatory state. IGF-1 inhibits the astrocytic response to inflammatory stimuli, and modulates microglial phenotype (IGF-1 promotes the microglial M2 and inhibits of M1 phenotype). Furthermore, IGF-1 is mitogenic for microglia. IGF-1 and estrogen interact to modulate the neuroinflammatory response and microglial and astrocytic phenotypes. Brain renin-angiotensin and IGF-1 systems also interact to modulate neuroinflammation. Induction of microglial IGF-1 by angiotensin, and possibly by other pro-inflammatory inducers, plays a major role in the repression of the M1 microglial neurotoxic phenotype and the enhancement of the transition to an M2 microglial repair/regenerative phenotype. This mechanism is impaired in aged brains. Aging-related decrease in IGF-1 may contribute to the loss of capacity of microglia to undergo M2 activation. Fine tuning of IGF-1 levels may be critical for regulating the neuroinflammatory response, and IGF-1 may be involved in inflammation in a context-dependent mode.
Collapse
Affiliation(s)
- Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Maria A Costa-Besada
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Carmen M Labandeira
- Department of Clinical Neurology, Hospital Alvaro Cunqueiro, University Hospital Complex, Vigo, Spain
| | - Begoña Villar-Cheda
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana I Rodríguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
40
|
Abstract
BACKGROUND Glaucoma is the second leading cause of blindness worldwide and is usually diagnosed in higher age groups. The goal was to survey how patient age influences the development of glaucoma. MATERIALS AND METHODS A web-based search on aging of the visual system and its influence on glaucoma was performed and the most important results are summarized. RESULTS The prevalence of glaucoma rises with age. Aging processes of the trabecular meshwork and the uveoscleral outflow pathway lead to a rise in the intraocular pressure. Chronically elevated intraocular pressure leads to remodelling of the lamina cribrosa and narrowing of its pores through which ganglion cell axons leave the eye. Age-dependent glia cell, mitochondria and immune system alterations are discussed to influence glaucoma. Patient age and further age-related nonophthalmological systemic diseases also influence adherence and persistence to the prescribed therapy. CONCLUSIONS Aging is an important risk factor for developing glaucoma and is a main factor which influences therapy and course of the disease. At this point in time it remains unclear to which extent additional factors determine the development of glaucoma.
Collapse
|
41
|
Labandeira-Garcia JL, Rodríguez-Perez AI, Garrido-Gil P, Rodriguez-Pallares J, Lanciego JL, Guerra MJ. Brain Renin-Angiotensin System and Microglial Polarization: Implications for Aging and Neurodegeneration. Front Aging Neurosci 2017; 9:129. [PMID: 28515690 PMCID: PMC5413566 DOI: 10.3389/fnagi.2017.00129] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022] Open
Abstract
Microglia can transform into proinflammatory/classically activated (M1) or anti-inflammatory/alternatively activated (M2) phenotypes following environmental signals related to physiological conditions or brain lesions. An adequate transition from the M1 (proinflammatory) to M2 (immunoregulatory) phenotype is necessary to counteract brain damage. Several factors involved in microglial polarization have already been identified. However, the effects of the brain renin-angiotensin system (RAS) on microglial polarization are less known. It is well known that there is a “classical” circulating RAS; however, a second RAS (local or tissue RAS) has been observed in many tissues, including brain. The locally formed angiotensin is involved in local pathological changes of these tissues and modulates immune cells, which are equipped with all the components of the RAS. There are also recent data showing that brain RAS plays a major role in microglial polarization. Level of microglial NADPH-oxidase (Nox) activation is a major regulator of the shift between M1/proinflammatory and M2/immunoregulatory microglial phenotypes so that Nox activation promotes the proinflammatory and inhibits the immunoregulatory phenotype. Angiotensin II (Ang II), via its type 1 receptor (AT1), is a major activator of the NADPH-oxidase complex, leading to pro-oxidative and pro-inflammatory effects. However, these effects are counteracted by a RAS opposite arm constituted by Angiotensin II/AT2 receptor signaling and Angiotensin 1–7/Mas receptor (MasR) signaling. In addition, activation of prorenin-renin receptors may contribute to activation of the proinflammatory phenotype. Aged brains showed upregulation of AT1 and downregulation of AT2 receptor expression, which may contribute to a pro-oxidative pro-inflammatory state and the increase in neuron vulnerability. Several recent studies have shown interactions between the brain RAS and different factors involved in microglial polarization, such as estrogens, Rho kinase (ROCK), insulin-like growth factor-1 (IGF-1), tumor necrosis factor α (TNF)-α, iron, peroxisome proliferator-activated receptor gamma, and toll-like receptors (TLRs). Metabolic reprogramming has recently been involved in the regulation of the neuroinflammatory response. Interestingly, we have recently observed a mitochondrial RAS, which is altered in aged brains. In conclusion, dysregulation of brain RAS plays a major role in aging-related changes and neurodegeneration by exacerbation of oxidative
stress (OS) and neuroinflammation, which may be attenuated by pharmacological manipulation of RAS components.
Collapse
Affiliation(s)
- Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de CompostelaSantiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED)Madrid, Spain
| | - Ana I Rodríguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de CompostelaSantiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED)Madrid, Spain
| | - Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de CompostelaSantiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED)Madrid, Spain
| | - Jannette Rodriguez-Pallares
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de CompostelaSantiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED)Madrid, Spain
| | - Jose L Lanciego
- Networking Research Center on Neurodegenerative Diseases (CIBERNED)Madrid, Spain.,Neurosciences Division, Center for Applied Medical Research (CIMA), University of NavarraPamplona, Spain
| | - Maria J Guerra
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de CompostelaSantiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED)Madrid, Spain
| |
Collapse
|
42
|
Ntsapi C, Loos B. Caloric restriction and the precision-control of autophagy: A strategy for delaying neurodegenerative disease progression. Exp Gerontol 2016; 83:97-111. [DOI: 10.1016/j.exger.2016.07.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 07/18/2016] [Accepted: 07/25/2016] [Indexed: 01/07/2023]
|
43
|
Yang J, Huang T, Song WM, Petralia F, Mobbs CV, Zhang B, Zhao Y, Schadt EE, Zhu J, Tu Z. Discover the network underlying the connections between aging and age-related diseases. Sci Rep 2016; 6:32566. [PMID: 27582315 PMCID: PMC5007654 DOI: 10.1038/srep32566] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 08/11/2016] [Indexed: 12/27/2022] Open
Abstract
Although our knowledge of aging has greatly expanded in the past decades, it remains elusive why and how aging contributes to the development of age-related diseases (ARDs). In particular, a global mechanistic understanding of the connections between aging and ARDs is yet to be established. We rely on a network modelling named "GeroNet" to study the connections between aging and more than a hundred diseases. By evaluating topological connections between aging genes and disease genes in over three thousand subnetworks corresponding to various biological processes, we show that aging has stronger connections with ARD genes compared to non-ARD genes in subnetworks corresponding to "response to decreased oxygen levels", "insulin signalling pathway", "cell cycle", etc. Based on subnetwork connectivity, we can correctly "predict" if a disease is age-related and prioritize the biological processes that are involved in connecting to multiple ARDs. Using Alzheimer's disease (AD) as an example, GeroNet identifies meaningful genes that may play key roles in connecting aging and ARDs. The top modules identified by GeroNet in AD significantly overlap with modules identified from a large scale AD brain gene expression experiment, supporting that GeroNet indeed reveals the underlying biological processes involved in the disease.
Collapse
Affiliation(s)
- Jialiang Yang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Tao Huang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Won-min Song
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Francesca Petralia
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Charles V. Mobbs
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Bin Zhang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Yong Zhao
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Eric E. Schadt
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Jun Zhu
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Zhidong Tu
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| |
Collapse
|
44
|
Garg G, Singh S, Singh AK, Rizvi SI. Metformin Alleviates Altered Erythrocyte Redox Status During Aging in Rats. Rejuvenation Res 2016; 20:15-24. [PMID: 27185159 DOI: 10.1089/rej.2016.1826] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Metformin, a biguanide drug commonly used to treat type 2 diabetes, has been noted to function as a caloric restriction mimetic. Its antidiabetic effect notwithstanding, metformin is currently being considered an antiaging drug candidate, although the molecular mechanisms have not yet been unequivocally established. This study aims to examine whether short-term metformin treatment can provide protective effects against oxidative stress in young and old-age rats. Young (age 4 months) and old (age 24 months) male Wistar rats were treated with metformin (300 mg/kg b.w.) for 4 weeks. At the end of the treatment period, an array of biomarkers of oxidative stress were evaluated, including plasma antioxidant capacity measured in terms of ferric reducing ability of plasma (FRAP), reactive oxygen species (ROS), lipid peroxidation (MDA), reduced glutathione (GSH), total plasma thiol (SH), plasma membrane redox system (PMRS), protein carbonyl (PCO), advanced oxidation protein products (AOPPs), and advanced glycation end products (AGEs) in control and experimental groups. Metformin treatment resulted in an increase in FRAP, GSH, SH, and PMRS activities in both age groups compared to respective controls. On the other hand, treated groups exhibited significant reductions in ROS, MDA, PCO, AOPP, and AGE level. Save for FRAP and protein carbonyl, the effect of metformin on all other parameters was more pronounced in old-aged rats. Metformin caused a significant increase in the PMRS activity in young rats, however, the effect was less pronounced in old rats. These findings provide evidence with respect to restoration of antioxidant status in aged rats after short-term metformin treatment. The findings substantiate the putative antiaging role of metformin.
Collapse
Affiliation(s)
- Geetika Garg
- Department of Biochemistry, Faculty of Science, University of Allahabad , Allahabad, India
| | - Sandeep Singh
- Department of Biochemistry, Faculty of Science, University of Allahabad , Allahabad, India
| | - Abhishek Kumar Singh
- Department of Biochemistry, Faculty of Science, University of Allahabad , Allahabad, India
| | - Syed Ibrahim Rizvi
- Department of Biochemistry, Faculty of Science, University of Allahabad , Allahabad, India
| |
Collapse
|
45
|
Ashpole NM, Herron JC, Mitschelen MC, Farley JA, Logan S, Yan H, Ungvari Z, Hodges EL, Csiszar A, Ikeno Y, Humphrey MB, Sonntag WE. IGF-1 Regulates Vertebral Bone Aging Through Sex-Specific and Time-Dependent Mechanisms. J Bone Miner Res 2016; 31:443-54. [PMID: 26260312 PMCID: PMC4854536 DOI: 10.1002/jbmr.2689] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 07/11/2015] [Accepted: 07/26/2015] [Indexed: 02/06/2023]
Abstract
Advanced aging is associated with increased risk of bone fracture, especially within the vertebrae, which exhibit significant reductions in trabecular bone structure. Aging is also associated with a reduction in circulating levels of insulin-like growth factor (IGF-1). Studies have suggested that the reduction in IGF-1 compromises healthspan, whereas others report that loss of IGF-1 is beneficial because it increases healthspan and lifespan. To date, the effect of decreases in circulating IGF-1 on vertebral bone aging has not been thoroughly investigated. Here, we delineate the consequences of a loss of circulating IGF-1 on vertebral bone aging in male and female Igf(f/f) mice. IGF-1 was reduced at multiple specific time points during the mouse lifespan: early in postnatal development (crossing albumin-cyclic recombinase [Cre] mice with Igf(f/f) mice); and in early adulthood and in late adulthood using hepatic-specific viral vectors (AAV8-TBG-Cre). Vertebrae bone structure was analyzed at 27 months of age using micro-computed tomography (μCT) and quantitative bone histomorphometry. Consistent with previous studies, both male and female mice exhibited age-related reductions in vertebral bone structure. In male mice, reduction of circulating IGF-1 induced at any age did not diminish vertebral bone loss. Interestingly, early-life loss of IGF-1 in females resulted in a 67% increase in vertebral bone volume fraction, as well as increased connectivity density and increased trabecular number. The maintenance of bone structure in the early-life IGF-1-deficient females was associated with increased osteoblast surface and an increased ratio of osteoprotegerin/receptor-activator of NF-κB-ligand (RANKL) levels in circulation. Within 3 months of a loss of IGF-1, there was a 2.2-fold increase in insulin receptor expression within the vertebral bones of our female mice, suggesting that local signaling may compensate for the loss of circulating IGF-1. Together, these data suggest the age-related loss of vertebral bone density in females can be reduced by modifying circulating IGF-1 levels early in life.
Collapse
Affiliation(s)
- Nicole M Ashpole
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jacquelyn C Herron
- Department of Immunology/Rheumatology/Allergy Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Matthew C Mitschelen
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Julie A Farley
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sreemathi Logan
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Han Yan
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Erik L Hodges
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Yuji Ikeno
- Department of Pathology, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mary Beth Humphrey
- Department of Immunology/Rheumatology/Allergy Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Department of Veterans' Affairs, Oklahoma City, OK, USA
| | - William E Sonntag
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
46
|
Szabó D, Gee NR, Miklósi Á. Natural or pathologic? Discrepancies in the study of behavioral and cognitive signs in aging family dogs. J Vet Behav 2016. [DOI: 10.1016/j.jveb.2015.08.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
47
|
Gambini J, Gimeno-Mallench L, Inglés M, Olaso G, Abdelaziz KM, Avellana JA, Belenguer Á, Cruz R, Mas-Bargues C, Borras C, Viña J. [Identification of single nucleotide polymorphisms in centenarians]. Rev Esp Geriatr Gerontol 2015; 51:146-9. [PMID: 26541311 DOI: 10.1016/j.regg.2015.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 09/01/2015] [Accepted: 09/09/2015] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Longevity is determined by genetic and external factors, such as nutritional, environmental, social, etc. Nevertheless, when living conditions are optimal, longevity is determined by genetic variations between individuals. In a same population, with relative genotypic homogeneity, subtle changes in the DNA sequence affecting a single nucleotide can be observed. These changes, called single nucleotide polymorphisms (SNP) are present in 1-5% of the population. MATERIAL AND METHODS A total of 92 subjects were recruited, including 28 centenarians and 64 controls, in order to find SNP that maybe implicated in the extreme longevity, as in the centenarians. Blood samples were collected to isolate and amplify the DNA in order to perform the analysis of SPN by Axiom™ Genotyping of Affymetrix technology. Statistical analyses were performed using the Plink program and libraries SNPassoc and skatMeta. RESULTS Our results show 12 mutations with a p<.001, where 5 of these (DACH1, LOC91948, BTB16, NFIL3 y HDAC4) have regulatory functions of the expressions of others genes. CONCLUSIONS Therefore, these results suggest that the genetic variation between centenarians and controls occurs in five genes that are involved in the regulation of gene expression to adapt to environmental changes better than controls.
Collapse
Affiliation(s)
- Juan Gambini
- Departamento de Fisiología, Facultad de Medicina, Universitat de València/INCLIVA, Valencia, España
| | - Lucía Gimeno-Mallench
- Departamento de Fisiología, Facultad de Medicina, Universitat de València/INCLIVA, Valencia, España
| | - Marta Inglés
- Departamento de Fisioterapia, Facultad de Fisioterapia, Universitat de València, Valencia, España
| | - Gloria Olaso
- Departamento de Fisiología, Facultad de Medicina, Universitat de València/INCLIVA, Valencia, España
| | - Kheira Mohamed Abdelaziz
- Departamento de Fisiología, Facultad de Medicina, Universitat de València/INCLIVA, Valencia, España
| | | | - Ángel Belenguer
- Servicio de Geriatría, Hospital de la Ribera, Alzira, Valencia, España
| | - Raquel Cruz
- CIBERER - Grupo de Medicina Xenómica, Universidad de Santiago de Compostela, España
| | - Cristina Mas-Bargues
- Departamento de Fisiología, Facultad de Medicina, Universitat de València/INCLIVA, Valencia, España
| | - Consuelo Borras
- Departamento de Fisiología, Facultad de Medicina, Universitat de València/INCLIVA, Valencia, España
| | - José Viña
- Departamento de Fisiología, Facultad de Medicina, Universitat de València/INCLIVA, Valencia, España.
| |
Collapse
|
48
|
Anisimov VN, Popovich IG, Zabezhinski MA, Egormin PA, Yurova MN, Semenchenko AV, Tyndyk ML, Panchenko AV, Trashkov AP, Vasiliev AG, Khaitsev NV. Sex differences in aging, life span and spontaneous tumorigenesis in 129/Sv mice neonatally exposed to metformin. Cell Cycle 2015; 14:46-55. [PMID: 25483062 DOI: 10.4161/15384101.2014.973308] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The perinatal (prenatal and early neonatal) period is a critical stage for hypothalamic programming of sexual differentiation as well as for the development of energy and metabolic homeostasis. We hypothesized that neonatal treatment with antidiabetic drug biguanide metformin would positively modify regulation of growth hormone--IGF-1--insulin signaling pathway slowing down aging and improving cancer preventive patterns in rodents. To test this hypothesis male and female 129/Sv mice were s.c. injected with metformin (100 mg/kg) at the 3rd, 5th and 7th days after birth. Metformin-treated males consumed less food and water and their body weight was decreased as compared with control mice practically over their entire lifespan. There were no significant differences in age-related dynamics of food and water consumption in females and they were heavier than controls. The fraction of mice with regular estrous cycles decreased with age and demonstrated a tendency to decrease in the females neonatally treated with metformin. Neonatal exposure to metformin practically failed to change the extent of hormonal and metabolic parameters in blood serum of male and female mice. In males, neonatal metformin treatment significantly increased the mean life span (+20%, P < 0.05) and slightly increased the maximum life span (+3.5%). In females, the mean life span and median in metformin-treated groups were slightly decreased (-9.1% and -13.8% respectively, P > 0.05) in comparison to controls, whereas mean life span of last 10% survivors and maximum life span were the same as in controls. Almost half (45%) of control male mice and 71.8% male mice neonatally exposed to metformin survived up to 800 d of age, the same age was achieved by 54.3% of mice in control female group and 30% of metformin-treated females (P < 0.03). Thus, neonatal metformin exposure slows down aging and prolongs lifespan in male but not in female mice.
Collapse
|
49
|
Oh E, Miller RA, Thurmond DC. Syntaxin 4 Overexpression Ameliorates Effects of Aging and High-Fat Diet on Glucose Control and Extends Lifespan. Cell Metab 2015; 22:499-507. [PMID: 26331606 PMCID: PMC4560841 DOI: 10.1016/j.cmet.2015.07.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/23/2014] [Accepted: 07/27/2015] [Indexed: 10/23/2022]
Abstract
Indirect evidence suggests that improved insulin sensitivity may contribute to improved lifespan of mice in which aging has been slowed by mutations, drugs, or dietary means, even in stocks of mice that do not show signs of late-life diabetes. Peripheral responses to insulin can be augmented by overexpression of Syntaxin 4 (Syn4), a plasma-membrane-localized SNARE protein. We show here that Syn4 transgenic (Tg) mice with high level expression of Syn4 had a significant extension of lifespan (33% increase in median) and showed increased peripheral insulin sensitivity, even at ages where controls exhibited age-related insulin resistance. Moreover, skeletal muscle GLUT4 and islet insulin granule exocytosis processes were fully protected in Syn4 Tg mice challenged with a high-fat diet. Hence, high-level expressing Syn4 Tg mice may exert better glycemic control, which slows multiple aspects of aging and extends lifespan, even in non-diabetic mice.
Collapse
Affiliation(s)
- Eunjin Oh
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan School of Medicine, Ann Arbor, MI 48109-2200, USA
| | - Debbie C Thurmond
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
50
|
Abstract
Neural stem cells (NSCs) are found in two regions in the adult brain: the subgranular zone (SGZ) in the hippocampal dentate gyrus and the subventricular zone (SVZ) adjacent to the lateral ventricles. Similarly to other somatic stem cells, adult NSCs are found within specialized niches that are organized to facilitate NSC self-renewal. Alterations in stem-cell homeostasis can contribute to the consequences of neurodegenerative diseases, healthy ageing and tissue repair after damage. Insulin and the insulin-like growth factors (IGFs) function in stem-cell homeostasis across species. Studies in the mammalian central nervous system support essential roles for IGF and/or insulin signalling in NSC self-renewal, neurogenesis, cognition and sensory function through distinct ligand-receptor interactions. IGF-II is of particular interest as a result of its production by the choroid plexus and presence in cerebrospinal fluid (CSF). CSF regulates and supports the development, division and migration of cells in the adult brain and is required for NSC maintenance. In this Review, we discuss emerging data on the functions of IGF-II and IGF and/or insulin receptor signalling in the context of NSC regulation in the SVZ and SGZ. We also propose a model for IGF-II in which the choroid plexus is a major component of the NSC niche.
Collapse
Affiliation(s)
- Amber N Ziegler
- Department of Neurology &Neuroscience, New Jersey Medical School, Rutgers Biomedical &Health Sciences, Cancer Centre, 205 South Orange Avenue, Newark, NJ 07101, USA
| | - Steven W Levison
- Department of Neurology &Neuroscience, New Jersey Medical School, Rutgers Biomedical &Health Sciences, Cancer Centre, 205 South Orange Avenue, Newark, NJ 07101, USA
| | - Teresa L Wood
- Department of Neurology &Neuroscience, New Jersey Medical School, Rutgers Biomedical &Health Sciences, Cancer Centre, 205 South Orange Avenue, Newark, NJ 07101, USA
| |
Collapse
|