bims-minfam Biomed News
on Inflammation and metabolism in ageing and cancer
Issue of 2024‒08‒25
eighteen papers selected by
Ayesh Seneviratne, Western University



  1. Nature. 2024 Aug 22.
      
    Keywords:  Ageing; Cancer; Public health
    DOI:  https://doi.org/10.1038/d41586-024-02713-x
  2. Nat Rev Nephrol. 2024 Sep;20(9): 557
      
    DOI:  https://doi.org/10.1038/s41581-024-00879-1
  3. Cardiovasc Res. 2024 Aug 23. pii: cvae178. [Epub ahead of print]
      Due to its peculiar structure and function, the cardiovascular system is particularly vulnerable to the detrimental effects of ageing. Current knowledge about the molecular mechanisms of ageing revealed the processes actively promoting ageing, e.g. progressive telomeres shortening, and the mechanisms opposing it, e.g. endogenous production of antioxidant substances. This knowledge can be used to measure biological age at a cellular and molecular level and to interfere with it by pharmacological or non-pharmacological interventions. Biological ageing is determined by the simultaneous occurrence of independent hallmarks, which encompass a wide range of biological processes, from genomic changes to systemic inflammation and dysbiosis. This narrative review will summarize the role of ageing hallmarks in the cardiovascular system, how they can be measured and what are the possible interventions to counteract their effects.
    Keywords:  ageing hallmarks; autophagy; cardiovascular ageing; epigenetic clocks; inflamm-ageing; sirtuins
    DOI:  https://doi.org/10.1093/cvr/cvae178
  4. Nature. 2024 Aug 19.
      
    Keywords:  Ageing; Brain; Machine learning; Neuroscience
    DOI:  https://doi.org/10.1038/d41586-024-02692-z
  5. Nature. 2024 Aug;632(8026): 729-730
      
    Keywords:  Ageing; Genetics; Genomics; Medical research
    DOI:  https://doi.org/10.1038/d41586-024-02677-y
  6. Aging (Albany NY). 2024 Aug 18. 16
      Aging is currently viewed as a result of multiple biological processes that manifest themselves independently, reinforce each other and in their totality lead to the aged phenotype. Genetic and pharmaceutical approaches targeting specific underlying causes of aging have been used to extend the lifespan and healthspan of model organisms ranging from yeast to mammals. However, most interventions display only a modest benefit. This outcome is to be expected if we consider that even if one aging process is successfully treated, other aging pathways may remain intact. Hence solving the problem of aging may require targeting not one but many of its underlying causes at once. Here we review the challenges and successes of combination therapies aimed at increasing the lifespan of mammals and propose novel directions for their development. We conclude that both additive and synergistic effects on mammalian lifespan can be achieved by combining interventions that target the same or different hallmarks of aging. However, the number of studies in which multiple hallmarks were targeted simultaneously is surprisingly limited. We argue that this approach is as promising as it is understudied.
    Keywords:  aging; combination therapy; hallmarks; lifespan and healthspan; mouse; synergistic effect
    DOI:  https://doi.org/10.18632/aging.206078
  7. Prostaglandins Leukot Essent Fatty Acids. 2024 Aug 10. pii: S0952-3278(24)00028-0. [Epub ahead of print]202 102634
      Aging is associated with systemic, non-resolving inflammation and the accumulation of senescent cells. The resolution of inflammation (or inflammation-resolution) is in part mediated by the balance between specialized pro-resolving mediators (SPMs) and pro-inflammatory leukotrienes (LTs). Aged mice (i.e. 2 years of age) exhibit a significant decrease in the SPM:LT ratio in specific organs including the spleen, which suggests that this organ may exhibit heightened inflammation and may be particularly amenable to SPM therapy. Previous studies have shown that resolvin D1 (RvD1) is decreased in spleens of aged mice compared with young controls. Therefore, we asked whether treatment of RvD1 in aged mice would impact markers of cellular senescence in splenic macrophages, and downstream effects on splenic fibrosis, a hallmark of splenic aging. We found that in aged mice, both zymosan-elicited and splenic macrophages showed an increase in mRNA expression of inflammatory and eicosanoid biosynthesis genes and a dysregulation of genes involved in the cell cycle. Injections with RvD1 reversed these changes. Importantly, RvD1 also decreased splenic fibrosis, a hallmark of splenic aging. Our findings suggest that RvD1 treatment may limit several features of aging, including senescence and fibrosis in spleens from aged mice. Summary Aging is associated with systemic, low grade, non-resolving inflammation. The resolution of inflammation is in part mediated by the balance between specialized pro-resolving mediators (SPMs) and pro-inflammatory lipid mediators, like leukotrienes (LTs). A hallmark of aging is the accumulation of senescent cells that promote low grade inflammation by secreting pro-inflammatory cytokines and lipid mediators. Splenic macrophages contribute to systemic aging, and spleens of aged mice demonstrate decreased levels of the SPM called resolvin D1 (RvD1). Whether addition of RvD1 is protective in spleens of aged mice is unknown and is focus of this study. RvD1 treatment to aged mice led to decreased mRNA expression of markers of cellular senescence and inflammation in splenic macrophages compared with age-matched vehicle controls. Moreover, RvD1 decreased splenic fibrosis, which occurs due to persistent low-grade inflammation in aging. Promoting inflammation resolution with RvD1 thus limits macrophage senescence, pro-inflammatory signals and established splenic fibrosis in aging.
    Keywords:  Aging; Fibrosis; Resolvins; Senescence; Spleen
    DOI:  https://doi.org/10.1016/j.plefa.2024.102634
  8. Free Radic Biol Med. 2024 Aug 15. pii: S0891-5849(24)00605-1. [Epub ahead of print]
      Oxidative metabolism declines with aging in humans leading to multiple metabolic ailments and subsequent inflammation. In mice, there is evidence of age-related suppression of fatty acid oxidation and oxidative phosphorylation in the liver, heart, and muscles. Many interventions that extend healthy lifespan of mice have been developed, including genetic, pharmacological, and dietary interventions. In this article, we review the literature on oxidative metabolism changes in response to those interventions. We also discuss the molecular pathways that mediate those changes, and their potential as targets for future longevity interventions.
    DOI:  https://doi.org/10.1016/j.freeradbiomed.2024.08.018
  9. Lancet Neurol. 2024 Sep;pii: S1474-4422(24)00265-5. [Epub ahead of print]23(9): 858-859
      
    DOI:  https://doi.org/10.1016/S1474-4422(24)00265-5
  10. Brain Res. 2024 Aug 20. pii: S0006-8993(24)00419-0. [Epub ahead of print]1844 149165
      Alzheimer's disease (AD) is a devastating neurodegenerative disease characterized by abnormal accumulation of tau proteins and amyloid-β, leading to neuronal death and cognitive impairment. Recent studies have implicated aging pathways, including dysregulation of tau and cellular senescence in AD pathogenesis. In AD brains, tau protein, which normally stabilizes microtubules, becomes hyperphosphorylated and forms insoluble neurofibrillary tangles. These tau aggregates impair neuronal function and are propagated across the brain's neurocircuitry. Meanwhile, the number of senescent cells accumulating in the aging brain is rising, releasing a pro-inflammatory SASP responsible for neuroinflammation and neurodegeneration. This review explores potential therapeutic interventions for AD targeting tau protein and senescent cells, and tau -directed compounds, senolytics, eliminating senescent cells, and agents that modulate the SASP-senomodulators. Ultimately, a combined approach that incorporates tau-directed medications and targeted senescent cell-based therapies holds promise for reducing the harmful impact of AD's shared aging pathways.
    Keywords:  Aging Pathways; Alzheimer’s Disease; Neurodegeneration; Senescent Cells; Tau Proteins; Therapeutic Targets
    DOI:  https://doi.org/10.1016/j.brainres.2024.149165
  11. Front Neurosci. 2024 ;18 1434945
      Age-related neurodegenerative diseases, like Alzheimer's disease (AD), are challenging diseases for those affected with no cure and limited treatment options. Functional, human derived brain tissues that represent the diverse genetic background and cellular subtypes contributing to sporadic AD (sAD) are limited. Human stem cell derived brain organoids recapitulate some features of human brain cytoarchitecture and AD-like pathology, providing a tool for illuminating the relationship between AD pathology and neural cell dysregulation leading to cognitive decline. In this review, we explore current strategies for implementing brain organoids in the study of AD as well as the challenges associated with investigating age-related brain diseases using organoid models.
    Keywords:  APOE; Alzheimer’s disease; aging; amyloid beta; brain organoids; cerebral organoids; hyperphosphorylated tau; neurodegenerative diseases
    DOI:  https://doi.org/10.3389/fnins.2024.1434945
  12. Cell Syst. 2024 Aug 21. pii: S2405-4712(24)00206-0. [Epub ahead of print]15(8): 738-752.e5
      Cellular longevity is regulated by both genetic and environmental factors. However, the interactions of these factors in the context of aging remain largely unclear. Here, we formulate a mathematical model for dynamic glucose modulation of a core gene circuit in yeast aging, which not only guided the design of pro-longevity interventions but also revealed the theoretical principles underlying these interventions. We introduce the dynamical systems theory to capture two general means for promoting longevity-the creation of a stable fixed point in the "healthy" state of the cell and the "dynamic stabilization" of the system around this healthy state through environmental oscillations. Guided by the model, we investigate how both of these can be experimentally realized by dynamically modulating environmental glucose levels. The results establish a paradigm for theoretically analyzing the trajectories and perturbations of aging that can be generalized to aging processes in diverse cell types and organisms.
    Keywords:  aging; caloric restriction; computational modeling; dynamical systems theory; longevity; metabolism; quantitative biology; single-cell imaging; systems biology; time-lapse microscopy
    DOI:  https://doi.org/10.1016/j.cels.2024.07.007
  13. Eur Heart J. 2024 Aug 20. pii: ehae515. [Epub ahead of print]
      
    DOI:  https://doi.org/10.1093/eurheartj/ehae515
  14. Front Aging. 2024 ;5 1471233
      
    Keywords:  C. elegans; Klotho; aging; lifespan; mTOR; nanohistology; vascular aging
    DOI:  https://doi.org/10.3389/fragi.2024.1471233
  15. Front Cell Neurosci. 2024 ;18 1433747
      The Aryl hydrocarbon receptor (AHR) is a cytosolic receptor and ligand-activated transcription factor widely expressed across various cell types in the body. Its signaling is vital for host responses at barrier sites, regulating epithelial renewal, barrier integrity, and the activities of several types of immune cells. This makes AHR essential for various cellular responses during aging, especially those governing inflammation and immunity. In this review, we provided an overview of the mechanisms by which the AHR mediates inflammatory response at gut and brain level through signals from intestinal microbes. The age-related reduction of gut microbiota functions is perceived as a trigger of aberrant immune responses linking gut and brain inflammation to neurodegeneration. Thus, we explored gut microbiome impact on the nature and availability of AHR ligands and outcomes for several signaling pathways involved in neurodegenerative diseases and age-associated decline of brain functions, with an insight on Parkinson's and Alzheimer's diseases, the most common neurodegenerative diseases in the elderly. Specifically, we focused on microbial tryptophan catabolism responsible for the production of several AHR ligands. Perspectives for the development of microbiota-based interventions targeting AHR activity are presented for a healthy aging.
    Keywords:  AHR; Alzheimer’s disease; Parkinson’s disease; microbiota-gut-brain axis; neuroinflammation; tryptophan metabolism
    DOI:  https://doi.org/10.3389/fncel.2024.1433747
  16. Eur J Intern Med. 2024 Aug 17. pii: S0953-6205(24)00345-5. [Epub ahead of print]
      Obesity is a disease that is assuming pandemic proportions in recent decades. With the advancement of medicine and increased access to care, average survival has increased, resulting in a larger number of elderly people. As a result, the amount of elderly people living with obesity is increasing, and the morbidity and impact of obesity on ageing implies severe limitations for these people. The link between obesity and ageing is not only epidemiological, but also strictly pathophysiological. Obesity accelerates the ageing process and ageing is characterised by pathophysiological mechanisms shared by obesity itself. Some examples of alterations shared by ageing and obesity are metabolic changes, sarcopenia and reduced functional capacity related to both loss of muscle strength and reduced cardiorespiratory fitness, as well as a general reduction in the perception of quality of life. The specific ability to antagonize these mechanisms through non-pharmacological treatment based on nutrition and exercise has always been one of the focal points of the international literature. Therefore, this review provides the state of the art on scientific knowledge regarding the main effects of an adequate nutritional plan and an individualised exercise prescription on the general health of elderly with obesity. In particular, this paper addresses the effect of nutrition and physical exercise on pathophysiological changes peculiar of ageing and obesity, providing also the scientific rational for nutritional and exercise prescription in the population.
    Keywords:  Aging; Body composition; Exercise prescription; Functional capacity; Nutrition; Protein intake
    DOI:  https://doi.org/10.1016/j.ejim.2024.08.007