Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters








Publication year range
1.
Trends Genet ; 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39341687

ABSTRACT

This review comprehensively examines the molecular biology and genetic origins of cellular senescence. We focus on various cellular stressors and pathways leading to senescence, including recent advances in the understanding of the genetic influences driving senescence, such as telomere attrition, chemotherapy-induced DNA damage, pathogens, oncogene activation, and cellular and metabolic stress. This review also highlights the complex interplay of various signaling and metabolic pathways involved in cellular senescence and provides insights into potential therapeutic targets for aging-related diseases. Furthermore, this review outlines future research directions to deepen our understanding of senescence biology and develop effective interventions targeting senescent cells (SnCs).

2.
bioRxiv ; 2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37425781

ABSTRACT

Combined multi-omics analysis of proteomics, polar metabolomics, and lipidomics requires separate liquid chromatography-mass spectrometry (LC-MS) platforms for each omics layer. This requirement for different platforms limits throughput and increases costs, preventing the application of mass spectrometry-based multi-omics to large scale drug discovery or clinical cohorts. Here, we present an innovative strategy for simultaneous multi-omics analysis by direct infusion (SMAD) using one single injection without liquid chromatography. SMAD allows quantification of over 9,000 metabolite m/z features and over 1,300 proteins from the same sample in less than five minutes. We validated the efficiency and reliability of this method and then present two practical applications: mouse macrophage M1/M2 polarization and high throughput drug screening in human 293T cells. Finally, we demonstrate relationships between proteomic and metabolomic data are discovered by machine learning.

3.
FEBS J ; 290(17): 4163-4186, 2023 09.
Article in English | MEDLINE | ID: mdl-35727858

ABSTRACT

Obesity and ageing predispose to numerous, yet overlapping chronic diseases. For example, metabolic abnormalities, including insulin resistance (IR) and type 2 diabetes (T2D) are important causes of morbidity and mortality. Low-grade chronic inflammation of tissues, such as the liver, visceral adipose tissue and neurological tissues, is considered a significant contributor to these chronic diseases. Thus, it is becoming increasingly important to understand what drives this inflammation in affected tissues. Recent evidence, especially in the context of obesity, suggests that the intestine plays an important role as the gatekeeper of inflammatory stimuli that ultimately fuels low-grade chronic tissue inflammation. In addition to metabolic diseases, abnormalities in the intestinal mucosal barrier have been linked to a range of other chronic inflammatory conditions, such as neurodegeneration and ageing. The flow of inflammatory stimuli from the gut is in part controlled by local immunological inputs impacting the intestinal barrier. Here, we will review the impact of obesity and ageing on the intestinal immune system and its downstream consequences on gut barrier function, which is strongly implicated in the pathogenesis of obesity and age-related diseases. In particular, we will discuss the effects of age-related intestinal dysfunction on neurodegenerative diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Obesity/metabolism , Inflammation , Aging , Liver/metabolism
4.
Nat Aging ; 2(8): 742-755, 2022 08.
Article in English | MEDLINE | ID: mdl-37118134

ABSTRACT

Cellular senescence is an important factor in aging and many age-related diseases, but understanding its role in health is challenging due to the lack of exclusive or universal markers. Using neural networks, we predict senescence from the nuclear morphology of human fibroblasts with up to 95% accuracy, and investigate murine astrocytes, murine neurons, and fibroblasts with premature aging in culture. After generalizing our approach, the predictor recognizes higher rates of senescence in p21-positive and ethynyl-2'-deoxyuridine (EdU)-negative nuclei in tissues and shows an increasing rate of senescent cells with age in H&E-stained murine liver tissue and human dermal biopsies. Evaluating medical records reveals that higher rates of senescent cells correspond to decreased rates of malignant neoplasms and increased rates of osteoporosis, osteoarthritis, hypertension and cerebral infarction. In sum, we show that morphological alterations of the nucleus can serve as a deep learning predictor of senescence that is applicable across tissues and species and is associated with health outcomes in humans.


Subject(s)
Aging, Premature , Deep Learning , Humans , Mice , Animals , Cellular Senescence/physiology , Aging , Biomarkers
5.
Nat Aging ; 1(9): 769-782, 2021 09.
Article in English | MEDLINE | ID: mdl-34746804

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic is a global health threat with particular risk for severe disease and death in older adults and in adults with age-related metabolic and cardiovascular disease. Recent advances in the science of ageing have highlighted how ageing pathways control not only lifespan but also healthspan, the healthy years of life. Here, we discuss the ageing immune system and its ability to respond to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We specifically focus on the intersect of severe COVID-19 and immunosenescence to highlight pathways that may be determinant for the risk of complications and death following infection with SARS-CoV-2. New or adapted therapeutics that target ageing-associated pathways may be important tools to reduce the burden of death and long-term disability caused by this pandemic. Proposed interventions aimed at immunosenescence could enhance immune function not only in the elderly but in susceptible younger individuals as well, ultimately improving complications of severe COVID-19 for all ages.


Subject(s)
Aging , COVID-19 , Immunosenescence , Humans , COVID-19/immunology , Immune System , SARS-CoV-2
7.
Nat Rev Mol Cell Biol ; 22(2): 119-141, 2021 02.
Article in English | MEDLINE | ID: mdl-33353981

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is a coenzyme for redox reactions, making it central to energy metabolism. NAD+ is also an essential cofactor for non-redox NAD+-dependent enzymes, including sirtuins, CD38 and poly(ADP-ribose) polymerases. NAD+ can directly and indirectly influence many key cellular functions, including metabolic pathways, DNA repair, chromatin remodelling, cellular senescence and immune cell function. These cellular processes and functions are critical for maintaining tissue and metabolic homeostasis and for healthy ageing. Remarkably, ageing is accompanied by a gradual decline in tissue and cellular NAD+ levels in multiple model organisms, including rodents and humans. This decline in NAD+ levels is linked causally to numerous ageing-associated diseases, including cognitive decline, cancer, metabolic disease, sarcopenia and frailty. Many of these ageing-associated diseases can be slowed down and even reversed by restoring NAD+ levels. Therefore, targeting NAD+ metabolism has emerged as a potential therapeutic approach to ameliorate ageing-related disease, and extend the human healthspan and lifespan. However, much remains to be learnt about how NAD+ influences human health and ageing biology. This includes a deeper understanding of the molecular mechanisms that regulate NAD+ levels, how to effectively restore NAD+ levels during ageing, whether doing so is safe and whether NAD+ repletion will have beneficial effects in ageing humans.


Subject(s)
Aging , Cell Physiological Phenomena , Metabolic Networks and Pathways , Mitochondria/metabolism , NAD/metabolism , Animals , Energy Metabolism , Humans
8.
Nat Metab ; 2(11): 1265-1283, 2020 11.
Article in English | MEDLINE | ID: mdl-33199924

ABSTRACT

Declining tissue nicotinamide adenine dinucleotide (NAD) levels are linked to ageing and its associated diseases. However, the mechanism for this decline is unclear. Here, we show that pro-inflammatory M1-like macrophages, but not naive or M2 macrophages, accumulate in metabolic tissues, including visceral white adipose tissue and liver, during ageing and acute responses to inflammation. These M1-like macrophages express high levels of the NAD-consuming enzyme CD38 and have enhanced CD38-dependent NADase activity, thereby reducing tissue NAD levels. We also find that senescent cells progressively accumulate in visceral white adipose tissue and liver during ageing and that inflammatory cytokines secreted by senescent cells (the senescence-associated secretory phenotype, SASP) induce macrophages to proliferate and express CD38. These results uncover a new causal link among resident tissue macrophages, cellular senescence and tissue NAD decline during ageing and offer novel therapeutic opportunities to maintain NAD levels during ageing.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Aging/metabolism , Cellular Senescence , Macrophage Activation , Membrane Glycoproteins/genetics , NAD/metabolism , ADP-ribosyl Cyclase/metabolism , Adipose Tissue, White/metabolism , Animals , Antigens, CD/metabolism , Cytokines/metabolism , Female , GPI-Linked Proteins/metabolism , Gene Expression , Glycolysis/genetics , Humans , Liver/metabolism , Male , Metabolome , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , NAD+ Nucleosidase/metabolism
9.
Cell Metab ; 26(3): 547-557.e8, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28877458

ABSTRACT

Ketogenic diets recapitulate certain metabolic aspects of dietary restriction such as reliance on fatty acid metabolism and production of ketone bodies. We investigated whether an isoprotein ketogenic diet (KD) might, like dietary restriction, affect longevity and healthspan in C57BL/6 male mice. We find that Cyclic KD, KD alternated weekly with the Control diet to prevent obesity, reduces midlife mortality but does not affect maximum lifespan. A non-ketogenic high-fat diet (HF) fed similarly may have an intermediate effect on mortality. Cyclic KD improves memory performance in old age, while modestly improving composite healthspan measures. Gene expression analysis identifies downregulation of insulin, protein synthesis, and fatty acid synthesis pathways as mechanisms common to KD and HF. However, upregulation of PPARα target genes is unique to KD, consistent across tissues, and preserved in old age. In all, we show that a non-obesogenic ketogenic diet improves survival, memory, and healthspan in aging mice.


Subject(s)
Aging/physiology , Diet, Ketogenic , Memory/physiology , Mortality , Animals , Diet, High-Fat , Fasting , Gene Expression Regulation , Liver/metabolism , Male , Mice, Inbred C57BL , PPAR alpha/genetics , PPAR alpha/metabolism
10.
Elife ; 52016 Feb 19.
Article in English | MEDLINE | ID: mdl-26894960

ABSTRACT

Macrophage activation/polarization to distinct functional states is critically supported by metabolic shifts. How polarizing signals coordinate metabolic and functional reprogramming, and the potential implications for control of macrophage activation, remains poorly understood. Here we show that IL-4 signaling co-opts the Akt-mTORC1 pathway to regulate Acly, a key enzyme in Ac-CoA synthesis, leading to increased histone acetylation and M2 gene induction. Only a subset of M2 genes is controlled in this way, including those regulating cellular proliferation and chemokine production. Moreover, metabolic signals impinge on the Akt-mTORC1 axis for such control of M2 activation. We propose that Akt-mTORC1 signaling calibrates metabolic state to energetically demanding aspects of M2 activation, which may define a new role for metabolism in supporting macrophage activation.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Macrophage Activation , Macrophages/metabolism , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Acetylation , Animals , Cell Proliferation , Chemokines/metabolism , Gene Expression Regulation , Histones/metabolism , Interleukin-4/metabolism , Macrophages/physiology , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , Protein Processing, Post-Translational
11.
Semin Immunol ; 27(4): 286-96, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26360589

ABSTRACT

Macrophages are pleiotropic cells that assume a variety of functions depending on their tissue of residence and tissue state. They maintain homeostasis as well as coordinate responses to stresses such as infection and metabolic challenge. The ability of macrophages to acquire diverse, context-dependent activities requires their activation (or polarization) to distinct functional states. While macrophage activation is well understood at the level of signal transduction and transcriptional regulation, the metabolic underpinnings are poorly understood. Importantly, emerging studies indicate that metabolic shifts play a pivotal role in control of macrophage activation and acquisition of context-dependent effector activities. The signals that drive macrophage activation impinge on metabolic pathways, allowing for coordinate control of macrophage activation and metabolism. Here we discuss how mTOR and Akt, major metabolic regulators and targets of such activation signals, control macrophage metabolism and activation. Dysregulated macrophage activities contribute to many diseases, including infectious, inflammatory, and metabolic diseases and cancer, thus a better understanding of metabolic control of macrophage activation could pave the way to the development of new therapeutic strategies.


Subject(s)
Macrophage Activation , Macrophages/metabolism , Signal Transduction , Animals , Glutamine/metabolism , Humans , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
12.
Cell Metab ; 19(6): 898-9, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24896536

ABSTRACT

Interleukin-6 (IL-6) is a pleiotropic cytokine that exerts either proinflammatory or anti-inflammatory effects and is implicated in diverse settings, including obesity, exercise, arthritis, and colitis. A new study shows that modulation of macrophage activation by IL-6 maintains glucose homeostasis in diet-induced obesity while limiting inflammation in endotoxemia (Mauer et al., 2014).


Subject(s)
Endotoxemia/immunology , Insulin Resistance , Interleukin-6/metabolism , Macrophage Activation , Macrophages/immunology , Obesity/immunology , Animals , Humans
13.
Nat Commun ; 4: 2834, 2013.
Article in English | MEDLINE | ID: mdl-24280772

ABSTRACT

Macrophages are able to polarize to proinflammatory M1 or alternative M2 states with distinct phenotypes and physiological functions. How metabolic status regulates macrophage polarization remains not well understood, and here we examine the role of mTOR (mechanistic target of rapamycin), a central metabolic pathway that couples nutrient sensing to regulation of metabolic processes. Using a mouse model in which myeloid lineage-specific deletion of Tsc1 (Tsc1(Δ/Δ)) leads to constitutive mTOR complex 1 (mTORC1) activation, we find that Tsc1(Δ/Δ) macrophages are refractory to IL-4-induced M2 polarization, but produce increased inflammatory responses to proinflammatory stimuli. Moreover, mTORC1-mediated downregulation of Akt signalling critically contributes to defective polarization. These findings highlight a key role for the mTOR pathway in regulating macrophage polarization, and suggest how nutrient sensing and metabolic status could be 'hard-wired' to control of macrophage function, with broad implications for regulation of type 2 immunity, inflammation and allergy.


Subject(s)
Cell Polarity/physiology , Macrophages/physiology , TOR Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Animals , Chitin , Inflammation Mediators/physiology , Interleukin-4/metabolism , Lipopolysaccharides/toxicity , Male , Mice , PPAR gamma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT6 Transcription Factor/metabolism , Tuberous Sclerosis Complex 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL