21 research outputs found

    Neuronal cell-based high-throughput screen for enhancers of mitochondrial function reveals luteolin as a modulator of mitochondria-endoplasmic reticulum coupling

    Get PDF
    Background: Mitochondrial dysfunction is a common feature of aging, neurodegeneration, and metabolic diseases. Hence, mitotherapeutics may be valuable disease modifiers for a large number of conditions. In this study, we have set up a large-scale screening platform for mitochondrial-based modulators with promising therapeutic potential. Results: Using differentiated human neuroblastoma cells, we screened 1200 FDA-approved compounds and identified 61 molecules that significantly increased cellular ATP without any cytotoxic effect. Following dose response curve-dependent selection, we identified the flavonoid luteolin as a primary hit. Further validation in neuronal models indicated that luteolin increased mitochondrial respiration in primary neurons, despite not affecting mitochondrial mass, structure, or mitochondria-derived reactive oxygen species. However, we found that luteolin increased contacts between mitochondria and endoplasmic reticulum (ER), contributing to increased mitochondrial calcium (Ca2+) and Ca2+-dependent pyruvate dehydrogenase activity. This signaling pathway likely contributed to the observed effect of luteolin on enhanced mitochondrial complexes I and II activities. Importantly, we observed that increased mitochondrial functions were dependent on the activity of ER Ca2+-releasing channels inositol 1,4,5-trisphosphate receptors (IP3Rs) both in neurons and in isolated synaptosomes. Additionally, luteolin treatment improved mitochondrial and locomotory activities in primary neurons and Caenorhabditis elegans expressing an expanded polyglutamine tract of the huntingtin protein. Conclusion: We provide a new screening platform for drug discovery validated in vitro and ex vivo. In addition, we describe a novel mechanism through which luteolin modulates mitochondrial activity in neuronal models with potential therapeutic validity for treatment of a variety of human diseases

    Brain energy rescue:an emerging therapeutic concept for neurodegenerative disorders of ageing

    Get PDF
    The brain requires a continuous supply of energy in the form of ATP, most of which is produced from glucose by oxidative phosphorylation in mitochondria, complemented by aerobic glycolysis in the cytoplasm. When glucose levels are limited, ketone bodies generated in the liver and lactate derived from exercising skeletal muscle can also become important energy substrates for the brain. In neurodegenerative disorders of ageing, brain glucose metabolism deteriorates in a progressive, region-specific and disease-specific manner — a problem that is best characterized in Alzheimer disease, where it begins presymptomatically. This Review discusses the status and prospects of therapeutic strategies for countering neurodegenerative disorders of ageing by improving, preserving or rescuing brain energetics. The approaches described include restoring oxidative phosphorylation and glycolysis, increasing insulin sensitivity, correcting mitochondrial dysfunction, ketone-based interventions, acting via hormones that modulate cerebral energetics, RNA therapeutics and complementary multimodal lifestyle changes

    The Aging Mitochondria

    No full text
    Mitochondrial dysfunction is a central event in many pathologies and contributes as well to age-related processes. However, distinguishing between primary mitochondrial dysfunction driving aging and a secondary mitochondrial impairment resulting from other cell alterations remains challenging. Indeed, even though mitochondria undeniably play a crucial role in aging pathways at the cellular and organismal level, the original hypothesis in which mitochondrial dysfunction and production of free radicals represent the main driving force of cell degeneration has been strongly challenged. In this review, we will first describe mitochondrial dysfunctions observed in aged tissue, and how these features have been linked to mitochondrial reactive oxygen species (ROS)–mediated cell damage and mitochondrial DNA (mtDNA) mutations. We will also discuss the clues that led to consider mitochondria as the starting point in the aging process, and how recent research has showed that the mitochondria aging axis represents instead a more complex and multifactorial signaling pathway. New working hypothesis will be also presented in which mitochondria are considered at the center of a complex web of cell dysfunctions that eventually leads to cell senescence and death

    Implication des interactions mitochondrie-réticulum endoplasmique dans le contrôle du métabolisme hépatique

    No full text
    Le foie est un organe indispensable dans le contrôle de l'homéostasie énergétique du corps humain. En particulier, le métabolisme hépatique est crucial pour l'homéostasie glucidique et lipidique. Les voies cataboliques et anaboliques sont en équilibre constant et régulées de façon synergique en fonction de la disponibilité en nutriments et de la demande en énergie. La perturbation de cet équilibre, notamment en cas d'obésité, peut conduire à l'accumulation intra-hépatique de lipides, qui est une des causes principales de la survenue de l'insulino-résistance hépatique (IRH), conduisant à l'hyperglycémie chronique et au diabète de type 2 (DT2). La cellule eucaryote est une structure hautement compartimentée, et à ce titre la compartimentalisation des processus cataboliques et anaboliques est une part intégrante de la gestion des voies métaboliques. Dans cet ensemble, la mitochondrie est un organite clef, qui abrite l'oxydation des lipides, le cycle de l'acide citrique (CAC) et la respiration cellulaire. De cette manière, la fonction mitochondriale est un élément crucial dans le maintien de l'état énergétique et d'oxydation-réduction de la cellule dans une gamme physiologique, ainsi que dans la régulation de l'activité du métabolisme du glucose et des lipides pour l'homéostasie du corps entier. La fonction mitochondriale est directement régulée par son interaction avec le réticulum endoplasmique (RE) via des zones de proximité entre les organites appelées Mitochondria-Associated-Endoplasmic-Reticulum-Membranes ou MAM. Dans ce contexte, j'ai participé au cours de mon travail de thèse à une étude qui a montré l'importance des interactions mitochondrie-RE dans la signalisation de l'insuline et mise en lumière la perturbation des MAM comme acteur principal dans l'IRH. De plus, j'ai étudié la régulation des MAM dans le contexte physiologique de la transition nutritionnelle dans le foie sain et insulino-résistant (IR)The liver is an essential organ in the control of energetic homeostasis of the human body. Particularly, hepatic metabolism is crucial for glucose and lipid homeostasis. Catabolism and anabolism of both substrates are in constant equilibrium and synergically regulated in regard of nutrient availability and energetic demand. Disruption of this equilibrium, especially in the case of obesity, can lead to hepatic accumulation of lipids, which is a major cause of hepatic insulin resistance (HIR) leading to chronic hyperglycaemia and type 2 diabetes (T2D). The eukaryotic cell is a highly compartmented structure, and in this respect compartmentation of anabolic and catabolic processes is an integral part of managing metabolic pathways together. In this context, the mitochondrion is a key organelle, housing oxidation of lipids, the tricarboxylic acid (TCA) cycle and cellular respiration. In this way, mitochondrial function is a crucial element in maintaining energetic and reductionoxidation state of the cell within physiological ranges, as well in regulating the proper activity of glucose and lipid metabolism for the all body homeostasis. Mitochondrial function is directly regulated by its interaction with the endoplasmic reticulum (ER) via proximity points between the organelles called Mitochondria-Associated-ER-Membranes (MAM). In this context I have participated during my Ph.D. in a work that has shown the importance of mitochondria-ER interactions in insulin signalling and highlighted MAM disruption as a main actor in HIR. Furthermore, I have studied the regulation of MAM in the physiological context of nutritional transition in the healthy and insulin resistant (IR) liver. Particularly, we have shown that MAM disruption induces impaired insulin signalling, while their reinforcement protects against its appearance and restore insulin sensitivity in lipid-induced IR condition. Moreover, we have pointed out a consistent decrease of MAM quantity in the IR liver of ob/ob, high-fat high-sucrose diet (HFHSD) and Cyclophilin D - knock-out (CypD-KO) mic

    The Aging Mitochondria

    No full text
    Mitochondrial dysfunction is a central event in many pathologies and contributes as well to age-related processes. However, distinguishing between primary mitochondrial dysfunction driving aging and a secondary mitochondrial impairment resulting from other cell alterations remains challenging. Indeed, even though mitochondria undeniably play a crucial role in aging pathways at the cellular and organismal level, the original hypothesis in which mitochondrial dysfunction and production of free radicals represent the main driving force of cell degeneration has been strongly challenged. In this review, we will first describe mitochondrial dysfunctions observed in aged tissue, and how these features have been linked to mitochondrial reactive oxygen species (ROS)-mediated cell damage and mitochondrial DNA (mtDNA) mutations. We will also discuss the clues that led to consider mitochondria as the starting point in the aging process, and how recent research has showed that the mitochondria aging axis represents instead a more complex and multifactorial signaling pathway. New working hypothesis will be also presented in which mitochondria are considered at the center of a complex web of cell dysfunctions that eventually leads to cell senescence and death

    Mitochondria-associated membranes response to nutrient availability and role in metabolic diseases

    No full text
    Metabolic diseases are associated with nutrient excess and metabolic inflexibility. Mitochondria and endoplasmic reticulum are important organelles and nutrient sensors, and their dysfunction has been extensively and independently implicated in metabolic diseases. Both organelles interact at sites known as mitochondria-associated membranes (MAMs), in order to exchange metabolites and calcium. Recent evidence indicates that MAM could be a hub of hepatic insulin signaling and nutrient sensing. In this review, we discuss the roles organelle function and communication play in the cell's adaptation to nutrient availability, in both physiology and metabolic diseases. We highlight how dynamic regulation of MAM affects mitochondria physiology and adaptation of cellular metabolism to nutrient availability, and how chronic MAM disruption participates in the metabolic inflexibility associated with metabolic disorders

    The Aging Mitochondria

    No full text

    Glucose dysregulation in pre-clinical Alzheimer's disease.

    No full text
    AD is the most common cause of age-related dementia and is consistently accompanied by glucose hypo-metabolism that precedes the onset of clinical symptoms, providing potential for early diagnosis and novel therapeutic interventions. Innovative inter-disciplinary research utilising appropriate pre-clinical models is required to further elucidate the molecular mechanisms, cross-talk with Aβ and tau pathology, and translatable approaches targeted to the early stages of AD. Given that glucose hypo‑metabolism has also been measured in other neurodegenerative diseases, we are excited to watch this field progress

    Reduced lactic acidosis risk with Imeglimin: Comparison with Metformin

    No full text
    International audienceThe global prevalence of type 2 diabetes (T2D) is expected to exceed 642 million people by 2040. Metformin is a widely used biguanide T2D therapy, associated with rare but serious events of lactic acidosis, in particular with predisposing conditions (e.g., renal failure or major surgery). Imeglimin, a recently approved drug, is the first in a new class (novel mode of action) of T2D medicines. Although not a biguanide, Imeglimin shares a chemical moiety with Metformin and also modulates mitochondrial complex I activity, a potential mechanism for Metformin-mediated lactate accumulation. We interrogated the potential for Imeglimin to induce lacticacidosis in relevant animal models and further assessed differences in key mechanisms known for Metformin's effects. In a dog model of major surgery, Metformin or Imeglimin (30-1000 mg/kg) was acutely administered, only Metformin-induced lactate accumulation and pH decrease leading to lactic acidosis with fatality at the highest dose. Rats with gentamycin-induced renal insufficiency received Metformin or Imeglimin (50-100 mg/kg/h), only Metformin increased lactatemia and H+ concentrations with mortality at higher doses. Plasma levels of Metformin and Imeglimin were similar in both models. Mice were chronically treated with Metformin or Imeglimin 200 mg/kg bid. Only Metformin produced hyperlactatemia after acute intraperitoneal glucose loading. Ex vivo measurements revealed higher mitochondrial complex I inhibition with Metformin versus slight effects with Imeglimin. Another mechanism implicated in Metformin's effects on lactate production was assessed: in isolated rat, liver mitochondria exposed to Imeglimin or Metformin, only Metformin (50-250 µM) inhibited the mitochondrial glycerol-3-phosphate dehydrogenase (mGPDH). In liver samples from chronically treated mice, measured mGPDH activity was lower with Metformin versus Imeglimin. These data indicate that the risk of lactic acidosis with Imeglimin treatment may be lower than with Metformin and confirm that the underlying mechanisms of action are distinct, supporting its potential utility for patients with predisposing conditions
    corecore