Friday, May 12, 2023
08h00-08h45
Registration & Documentation
Foyer do Salão Nobre FFUP
Collect your documentation & Mount your Poster
8h45-9h00
Welcome Address
Salão Nobre
8h45-9h00 Welcome address
9h00-10h30
Session I
Salão Nobre
Chairs
Vanessa Morais, Jorge MA Oliveira
9h00-10h00 Opening Lecture
Mitochondrial superoxide in ischemia-reperfusion injury
Mike Murphy (MRC Mitochondrial Biology Unit, University of Cambridge, United Kingdom)
Click for Abstract
Mitochondrial redox metabolism is central to the life and death of the cell. For example, mitochondrial production of free radicals and subsequent oxidative damage has long been known to contribute to damage in conditions such as ischaemia-reperfusion (IR) injury in stroke and heart attack. More recently mitochondrial redox changes have also been implicated in redox signalling. Over the past years we have developed a series of mitochondria-targeted compounds designed to ameliorate or determine how these changes occur. I will outline some of this work, which suggested that ROS production in IR injury during stroke was mainly coming from complex I. This led us to investigate the mechanism of the ROS production and using a metabolomic approach we found that the ROS production in IR injury came from the accumulation of succinate during ischaemia that then drove mitochondrial ROS production by reverse electron transport at complex I during reperfusion. This surprising mechanism led up to develop further new therapeutic approaches to impact on the damage that mitochondrial ROS do in pathology and also to explore how mitochondrial ROS can act as redox signals. I will discuss how these unexpected mechanisms may lead to redox and metabolic signals from mitochondria in a range of conditions under both healthy and pathological conditions.
References
A unifying mechanism for mitochondrial superoxide production during ischemia reperfusion injury
Edward T. Chouchani, Victoria R. Pell, Andrew M. James, Lorraine M. Work, Kourosh Saeb-Parsy, Christian Frezza, Thomas Krieg and Michael P. Murphy
Cell Metabolism (2016) 23 254-263
Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS
Edward T. Chouchani, Victoria R. Pell, Edoardo Gaude, Dunja Aksentijević, Stephanie Y. Sundier, Ellen L. Robb, Angela Logan, Sergiy M. Nadtochiy, Emily N. J. Ord, Anthony C. Smith, Filmon Eyassu, Rachel Shirley, Chou-Hui Hu, Anna J. Dare, Andrew M. James, Sebastian Rogatti, Richard C. Hartley, Simon Eaton, Ana, S. H. Costa, Paul S. Brookes, Sean M. Davidson, Michael R. Duchen, Kourosh Saeb-Parsy, Michael J. Shattock, Alan J. Robinson, Lorraine M. Work, Christian Frezza, Thomas Krieg and Michael P. Murphy
Nature (2014) 515 431-435
10h00-10h15
Mitochondrial DNA integrity defines muscle satellite cells determination into skeletal muscle fibres
Ayesha Sen (University of Cologne, Centre for Physiology and Pathophysiology, Germany)
Click for Abstract
Mitochondrial DNA integrity defines muscle satellite cells determination into skeletal muscle fibres
Ayesha Sen (1), Rudolf J. Wiesner (1,2,3), David Pla-Martín (1,2)
1- University Clinic Cologne, University of Cologne, Centre for Physiology and Pathophysiology
2- University of Cologne, CMMC
3-University of Cologne, CECAD
Mutations in the mitochondrial DNA (mtDNA) have been linked to skeletal muscle atrophy and sarcopenia. Muscle satellite cells (MuSCs), responsible for regenerating new muscle fibres, are generally quiescent but become activated during muscle biogenesis, damage or exercise. To explore how mtDNA mutations affect the regeneration capacity of MuSCs we employ a mouse model expressing a dominant negative mutation of TWINKLE (p.K320E), the mtDNA helicase, under the control of tamoxifen, specifically in the MuSCs. This mutation is known to accelerate accumulation of mtDNA alterations. In quiescent stem cells, K320E-Twinkle is well tolerated, and we only observe mtDNA depletion in very old mice. Activation of muscle regeneration in adult mice by cardiotoxin injection in the Tibialis Anterior, induces the accumulation of fibres with mitochondrial dysfunction, mtDNA alterations and a prominent fibre type shift. In contrast, if K320E-Twinkle is activated in a more physiological manner during muscle biogenesis, we do not observe mitochondrial dysfunction but, again, a prominent shift in fibre type. In these mice, cardiotoxin injection one year after K320E-Twinkle activation, did not result in accumulation of fibres with mitochondrial dysfunction. In addition, organelle proteomics of C2C12 expressing K320E-Twinkle revealed lower accumulation of mitochondrial proteins during fibre differentiation. We propose that the fibre type shift observed during ageing might be the consequence of the accumulation of mtDNA damage; a process that has been observed in all tissues of organisms with a long lifespan, including humans.
10h15-10h30
Overexpression of UCP4 in astrocytic mitochondria prevents multilevel dysfunctions in a mouse model of Alzheimer's disease
Jean-Yves Chatton (Dep. Fundamental Neurosciences, Univ. Lausanne, Switzerland)
Click for Abstract
Overexpression of UCP4 in astrocytic mitochondria prevents multilevel dysfunctions in a mouse model of Alzheimer's disease
Nadia Rosenberg (1), Maria Reva (2), Francesca Binda (1), Leonardo Restivo (1), Pauline Depierre (1), Julien Puyal (1), Marc Briquet (1), Yann Bernardinelli (3), Anne-Bérengère Rocher (1), Henry Markram (2), Jean-Yves Chatton (1)
1- Department of Fundamental Neurosciences,University of Lausanne, Lausanne, Switzerland
2- Blue Brain Project (BBP), Ecole polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
3- Neonomia, Geneva, Switzerland
Alzheimer's disease (AD) is becoming increasingly prevalent worldwide. It represents one of the greatest medical challenges as no pharmacologic treatments are available to prevent disease progression. Astrocytes play crucial functions within neuronal circuits by providing metabolic and functional support, regulating interstitial solute composition, and modulating synaptic transmission. In addition to these physiological functions, growing evidence points to an essential role of astrocytes in neurodegenerative diseases like AD. Early-stage AD is associated with hypometabolism and oxidative stress. Contrary to neurons that are vulnerable to oxidative stress, astrocytes are particularly resistant to mitochondrial dysfunction and are therefore more resilient cells. In our study, we leveraged astrocytic mitochondrial uncoupling and examined neuronal function in the 3xTg AD mouse model. We overexpressed the mitochondrial uncoupling protein 4 (UCP4), which has been shown to improve neuronal survival in vitro. We found that this treatment efficiently prevented alterations of hippocampal metabolite levels observed in AD mice, along with hippocampal atrophy and reduction of basal dendrite arborization of subicular neurons. This approach also averted aberrant neuronal excitability observed in AD subicular neurons and preserved episodic-like memory in AD mice assessed in a spatial recognition task. These findings show that targeting astrocytes and their mitochondria is an effective strategy to prevent the decline of neurons facing AD-related stress at the early stages of the disease.
10h30-11h15
Coffee-Break + Posters
Foyer
11h15-13h00
Session II
Salão Nobre
Chairs
Claúdia Pereira, Paulo J Oliveira
11h15 - 11h45
Mitochondrial DNA mutations in ageing and cancer – what’s the connection?
Laura Greaves (Wellcome Trust Centre for Mitochondrial Research, United Kingdom)
Click for Abstract
Alterations in mitochondrial metabolism are major hallmarks of both ageing cells and cancer. Age is the biggest risk factor for the development of a significant number of cancer types and this therefore raises the question of whether there is a link between age-related mitochondrial dysfunction and the advantageous changes in mitochondrial metabolism prevalent in cancer cells. A common underlying feature of both ageing and cancer cells is the presence of somatic mutations of the mitochondrial genome (mtDNA). MtDNA mutations are particularly enriched in colorectal cancers [1] and we have previously shown that individual normal human colonic crypt stem cells also accumulate somatic mtDNA point mutations with age [2]. This shows that somatic mtDNA mutations and altered metabolic pathways are present in colonic crypts prior to malignant transformation, suggesting that mtDNA mutations may either increase the risk of malignant transformation, promote tumour progression, or are selectively propagated during tumour development. To investigate this we generated a mouse model in which we induced tumours specifically in intestinal stem cells with and without mtDNA mutation-induced mitochondrial dysfunction. We found that the mice with mitochondrial dysfunction had similar numbers of tumours to controls but they were growing significantly faster resulting in a shortened lifespan [3]. Multi-omics analysis revealed the underlying mechanism to be an upregulation of the de novo serine synthesis pathway and mitochondrial one-carbon metabolism in response to mitochondrial dysfunction. These anabolic pathways are important regulators of cellular biomass production and, excitingly, may represent metabolic vulnerabilities for therapeutic exploitation in human colorectal cancer.
References
1 - Gorelick, A. N. et al. Nat Metab 3, 558-570, (2021).
2 - Greaves, L. C. et al. Exp Gerontol 45, 573-579, (2010).
3 - Smith, A. L. M. et al. Nature Cancer 1, 976-989, (2020).
11h45-12h00
Evolution and maintenance of mtDNA gene content across eukaryotes
Iain Johnston (University of Bergen, Norway)
Click for Abstract
Mitochondrial DNA (mtDNA) encodes essential cellular machinery in almost all eukaryotes. But the mitochondrion is an awkward place to store genetic information safely; it's a physically and chemically damaging environment involving frequent and error-prone replication. Most genes for mitochondrial machinery have been transferred to the nucleus since endosymbiosis. So why do mitochondria retain any genes at all? Why do different species retain dramatically different mitochondrial gene counts? And how do they mitigate against the inevitable damage in these essential genes?
I'll talk about our work combining bioinformatics, modelling, heteroplasmy profiling across taxa, and microscopy to test long-standing and new hypotheses about why mtDNA retains genes, and how species in different kingdoms and with different lifestyles can use mitochondrial structure and dynamics to segregate mtDNA damage. In organisms that cannot sequester a protected germline and so use an animal-like mtDNA bottleneck, I'll describe theoretical and experimental results showing that mtDNA recombination can serve this purpose -- at the cost of requiring innovative cellular behaviour to deal with the consequences.
References
Giannakis, K., Arrowsmith, S.J., Richards, L., Gasparini, S., Chustecki, J.M., Røyrvik, E.C. and Johnston, I.G., 2022. Evolutionary inference across eukaryotes identifies universal features shaping organelle gene retention. Cell Systems, 13(11), pp.874-884.
García Pascual, B., Nordbotten, J.M. and Johnston, I.G., 2023. Cellular and environmental dynamics influence species-specific extents of organelle gene retention. Proceedings of the Royal Society B, 290(1994), p.20222140.
Edwards, D.M., Røyrvik, E.C., Chustecki, J.M., Giannakis, K., Glastad, R.C., Radzvilavicius, A.L. and Johnston, I.G., 2021. Avoiding organelle mutational meltdown across eukaryotes with or without a germline bottleneck. PLoS biology, 19(4), p.e3001153.
Broz, A.K., Keene, A., Fernandes Gyorfy, M., Hodous, M., Johnston, I.G. and Sloan, D.B., 2022. Sorting of mitochondrial and plastid heteroplasmy in Arabidopsis is extremely rapid and depends on MSH1 activity. Proceedings of the National Academy of Sciences, 119(34), p.e2206973119.
Acknowledgements
This project has received funding from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation program (grant agreement no. 805046 [EvoConBiO]). Support from the Peder Sather fund is gratefully acknowleged.
12h00-12h15
Mitochondrial ROS as a regulator of synaptic function
Pablo Peixoto (Baruch College CUNY, New York, NY, USA)
Click for Abstract
Mitochondrial ROS as a regulator of synaptic function
Irina Stavrovskaya (1), Bethany Kristi Morin (2), Pablo Peixoto (1,2)
1- Baruch College CUNY, New York, NY, USA,
2- Graduate Center, CUNY, New York, NY, USA
Exacerbated emission of reactive oxygen species (ROS) from presynaptic mitochondria is a well-studied hallmark of several neurodegenerative diseases, including amyotrophic lateral sclerosis. Outside of the context of disease, the potential role of mitochondrial ROS in presynaptic function and plasticity remains largely understudied. Here, we investigated this potential role by combining electrophysiological recordings, confocal imaging, immunohistochemical staining and optogenetics using a well-established protocol for induction and measurement of synaptic potentiation in drosophila neuromuscular preparations. We have previously found that optogenetic induction of ROS emission from presynaptic mitochondria expressing mitokiller red was accompanied by an increase in spontaneous mini junction potentials. The increase in electrical activity did not appear to be associated
with major synaptic structure alterations such as the formation of presynaptic filopodia or the growth of ghost boutons. However, in existing boutons, we observed an increase in active zone markers such as nc82/Brp. We have not detected Wnt1/Wg release from synaptic boutons suggesting the involvement of other signaling pathways that underlie observed changes in electrophysiological activity mediated by ROS emission. Future studies will further inquire the role of mitochondrial ROS in synaptic potentiation, as well as the potential signaling targets of mitochondrial ROS in the presynaptic structure.
12h15-12h30 Sponsored Talk
Tebubio: A unique combination of solutions to facilitate deciphering of Mitochondria Biology
Frédéric Samazan (TebuBio, France)
Click for abstract
At Tebubio our vision of helping to advance Life Sciences research every day, is embodied by providing a unique, holistic range of solutions to make your life easier - simply and quickly accessible.
In order to match your requirements and provide the most suitable solution to facilitate all your projects to decipher Mitochondria biology , our experts provide reliable, multimodal solutions, encompassing:
• An extensive range of routine reagents (antibodies - ready to use assay kits - Mitochondria targeted compound libraries) to more innovative and customized solutions such as new live cell imaging probe to measure mitochondria membrane tension or customized Biological samples..
• An In-house laboratory services – to quickly and easily extend your ressources, and move your projects forward faster, with our ready-to-go or personalized solutions (from cell culture to biomarker discovery and customized mRNA production service).
With Tebubio you’ll find a global partner, with a unique combination of solutions and expertise enabling you to accelerate all your projects and to concentrate on what’s most important for you: Deciphering Mitochondria Biology.
12h30 - 13h00 FLASH TALKS (Each is a 5 min oral presentation, complemented with a Poster)
Imaging of mitochondrial properties of a living mouse brain using in vivo two-photon microscopy
Renata Couto (iMM - Institute of Molecular Medicine, Portugal)
Click for abstract
Imaging of mitochondrial properties of a living mouse brain using in vivo two-photon microscopy
Renata Couto (1), Miguel Remondes (1), Vanessa A. Morais (1)
1- iMM Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
Parkinson’s Disease (PD) is a neurodegenerative disorder with a multifactorial etiology, whose hallmarks include the build-up of misfolded protein aggregates termed Lewy bodies and specific loss of dopaminergic neurons from the substantia nigra pars compacta. Many risk factors have been linked with the establishment and progression of PD, including aging, exposure to environmental toxic agents, and genetic factors, all of which seem to particularly impact mitochondria. Among the known PD genetic mutations, alterations in the PTEN-induced kinase 1 (PINK1) gene have been identified in specific recessive familial PD forms. PINK1 is a mitochondrial serine/threonine kinase protein whose function relies on the mitochondrial membrane potential. In the presence of defective mitochondria, PINK1 coordinates the removal of defective organelles from the cell, whereas under basal conditions it is internalized and mediates the phosphorylation of the oxidative phosphorylation chain (OXPHOS) Complex I subunit NDUFA10, regulating the overall energetic output of the cell.
Even though defects in lipid metabolism and cellular bioenergetics have been found in PD patients and models, the precise mechanisms underlying these processes and the link with mitochondrial dysfunction remains to be clarified. In line with this, new evidences are also emerging on a role for PINK1 in the regulation of this metabolic processes. Of relevance, alterations in many mitochondrial-related enzymes implicated in the fatty acid oxidation (FAO) and lipid synthesis, have been linked with PINK1.
Recently, in a phosphoproteomic analysis performed by the host lab, ACAD9 was identified as a putative PINK1 substrate. ACAD9 is a mitochondrial-targeted protein involved in FAO and Complex I biogenesis, and even though ACAD9’s function in the cell has been described, its interacting counterparts remain to be clarified. Furthermore, whether ACAD9 and PINK1, two proteins implicated in lipid metabolism and Complex I regulation, are involved within the same pathway is still unknown.
Therefore, our goal is to validate whether ACAD9 is a PINK1 substrate and to understand the importance of this PINK1-ACAD9 axis for the regulation of lipid metabolism and Complex I function. Ultimately, we aim at clarifying whether the PINK1-ACAD9 axis has a central role in the neuronal homeostasis, in general, and in the pathophysiology of PD, in particular.
Antibiotics that cause mitochondrial dysfunction can inhibit the growth of tumors derived from chemoresistant or stem cancer cells
Alex Lyakhovich (Sabanci University, Istanbul, Turkey)
Click for abstract
Antibiotics that cause mitochondrial dysfunction can inhibit the growth of tumors derived from chemoresistant or stem cancer cells
Alex Lyakhovich (1), Cemile Uslu (1), Can Tunçay (1), Aslı Özge Özkul (2), Didenur Çevik (1), Ece Yücel (1), Eda Kapan (1), Zeynep Ülker (1), and Etna Abad (3)
1- Sabanci University, Istanbul, Turkey
2- Bahcesehir University, Istanbul, Turkey
3- Pompeu Fabra University, Barcelona, Spain
A subset of chemoresistant (CRC) and cancer stem cells (CSC) is thought to be responsible for metastatic disease and is a major cause of cancer-related mortality. Our recent observations suggest that both CRC and CSC have a similar spectrum of stemness markers as compared to the parental cancer cells from which they originated. In a triple-negative breast cancer models, we observed that CSC and CRC exhibited higher mitochondrial membrane potential, greater oxidative phosphorylation (OXPHOS) activity, and increased resistance to oxidative stress, enabling them to survive in harsh microenvironments such as during chemotherapy or radiation therapy. We also found increased expression of OXPHOS-associated proteins in these cells, indicating a potential target for cancer therapy. Our in vivo findings indicate that certain antibiotics that inhibit mitochondrial function can reduce the growth rate of tumors developed from CSC or CRC. To improve drug efficacy, we have modified antibiotics selected from the chemical library for specific delivery to the mitochondria. Inhibition of mitophagy can enhance this effect. In conclusion, our study highlights the potential of targeting the unique metabolic characteristics of CSC and CRC with antibiotics that inhibit mitochondrial function, providing an alternative avenue for anticancer therapy that may improve patient outcomes for those with resistant and metastatic tumors.
Breaking the Cycle: Exercise During Pregnancy Can Improve Cardiovascular Health in Offspring of High Fat, High Sugar-Fed Mothers
Mariana S. Diniz (CNC, CIBB Biocant, PDBEB, IIIUC, University of Coimbra, Portugal)
Click for abstract
Breaking the Cycle: Exercise During Pregnancy Can Improve Cardiovascular Health in Offspring of High Fat, High Sugar-Fed Mothers
Susana P. Pereira (1,2*), Mariana S. Diniz (1,3*), João D. Martins (1*), Óscar M. Rodrigues (1), Carolina Tocantins (1,3), Luís F. Grilo (1,3), Jelena Stevanovic-Silva (2), Jorge Beleza (2), Pedro Coxito (2), David Rizo-Roca (4), Estela Santos-Alves (2), Manoel Rios (2), António J. Moreno (1,5), António Ascensão (2), José Magalhães (2) and Paulo J. Oliveira (1)
* - Equal Contribution
1- CNC - Center for Neuroscience and Cell Biology, CIBB, Biocant Park, University of Coimbra, Coimbra, Portugal
2- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, Porto, Portugal
3- Ph.D. Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
4- Cellular Biology Department, Physiology and Immunology, Faculty of Biology, University of Barcelona, Spain
5- Life Sciences Department, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
Background: Maternal obesity (MO) during pregnancy creates an adverse intra-uterine environment, affecting fetal development and increasing cardiac disease risk in offspring. It is estimated that 40 million pregnant women are overweight or obese. Cardiac mitochondria are essential for energy dysfunction and their dysfunction is implicated in cardiovascular disease (CVD). Maternal physical exercise during obesity (MOEx) may mitigate MO-induced offspring’s CVD risks by improving mitochondrial function. However, it is unclear whether the effects of gestational exercise during MO on offspring cardiac mitochondria can be persistent in time.
Aim: Investigate whether MOEx induces adaptations in offspring’s cardiac mitochondrial bioenergetics that persist until young adulthood.
Methods: A Sprague-Dawley MO rat model was achieved by feeding a high-fat-high-sugar (HFHS) diet. Six HFHS-fed mothers were kept sedentary (MO; n=6), six exercised (MOEx; n=6), and other six received a control chow diet (C; n=6). Offspring were kept on a standard chow diet without exercise. Male and female offspring from each group (F1-C; F1-MO; F1-MOEx) were euthanized at 32 weeks old (n=6/sex) and cardiac tissue and blood plasma collected. Blood biochemical parameters were measured through mass spectrometry. Cardiac mitochondrial respiration was measured in vitro using Oxygraph, and membrane potential determined with TPP+-electrode. Protein expression was evaluated through Western blot. Unpaired t-test or Mann-Whitney statistical tests were applied (p≤0.05).
Results: In this study, offspring of mothers exercised during pregnancy (F1-MOEx) had lower triglyceride blood levels vs F1-MO in males (p<0.01) and females (p<0.01). The atherogenic index (AI), reflecting CVD risk, was increased in females F1-MO vs F1-C (p<0.01). AI was decreased in females F1-MOEx vs F1-MO (p<0.01) and males F1-MOEx vs F1-C (p=0.01). CD36 expression levels were decreased in males F1-MOEx vs F1-C (p=0.04). Males F1-MOEx showed increased expression levels of the mitochondrial complex(C)-II subunit SDHA vs F1-C (p=0.04). CII-supported RCR was increased in males F1-MOEx vs F1-C (p=0.01) and F1-MO (p<0.01). Females F1-MOEx showed shorter lag-phase for CII-supported ADP phosphorylation vs F1-C (p=0.01) and F1-MO (p=0.04).
Conclusions: This work suggests that MOEx can modulate offspring’s cardiac mitochondrial bioenergetics during fetal development in a way that persists until 32-weeks-old, in a sex-specific way, especially in mitochondrial CII. This may be favorable for improving offspring’s cardiovascular health by decreasing MO-induced increased CVD risk. This underscores the importance of exercise during MO and suggests that MOEx may mitigate MO adverse effects on offspring cardiovascular health.
Support: FCT Fellowships (SFRH/BPD/116061/2016; SFRH/BD/11934/2022;SFRH/BD/11924/2022;SFRH/BD/5539/2020), PTDC/DTP-DES/1082/2014 (POCI-01-0145-FEDER-016657), CENTRO-01-0246-FEDER 000010, UIDB/04539/2020, UIDP/04539/2020, LA/P/0058/2020
13h00 - 14h30
Lunch, Posters & Networking
14h30 - 16h00
Session III
Salão Nobre
Chairs
Laura Greaves, Jorge MA Oliveira
14h30 - 15h00
Mitochondrial disease heterogeneity: Contrasting phenotypes in two mouse models with mt-tRNA-Ala mutations
Jim Stewart (Wellcome Trust Centre for Mitochondrial Research, United Kingdom)
Click for Abstract
Diseases caused by mutations of the mitochondrial DNA show a surprisingly diverse array of variable physical manifestations, age of onset, and severity in patients – even for mutations within the same gene. This tissue and cell-specific variability in the disease presentation necessitates animal model research to understand these phenomena and lead to translational breakthroughs for mitochondrial disease patients. Despite advances in nuclear genome-engineering, animal mitochondrial-DNA has remained resistant to transgenic manipulation until quite recently. I will present out “phenotype first” method of screening for mouse models of heteroplasmic, pathological mtDNA mutations and contrast two models with a 5 bp difference between the mutational sites in the mt-tRNAAla gene.
15h00 - 15h15
Mitochondrial DNA variation and stress responses
Aurora Gomez-Duran (Universidade de Santiago de Compostela, Spain)
Mitochondrial DNA variation and stress responses
Aurora Gomez-Duran
Centro de Investigaciones Biológicas Margarita Salas. CSIC. 28040, Madrid, Spain
Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas. CiMUS. Universidade de Santiago de Compostela. 15782. A Coruña.
Mitochondrial DNA (mtDNA) variants influence the risk of rare and late-onset human diseases, but the reasons for this are poorly understood. Interestingly, the same variant exerts a great variability in disease penetrance in each individual, which suggests the existence of a complex system that does not necessarily imply the dysfunction of the energy synthesis. In here, through the combination of multi-omics approaches on several human models, we will describe how variations in oxidative phosphorylation system capacity (OXPHOS) driven by the mtDNA variants activate different types of stress responses and their possible role in late-onset disease. We will further show how these findings can be applied to pharmacogenomic discovery and search of new biomarkers.
15h15 - 15h30
MitoKO: A library of base editors for the precise ablation of all protein-coding genes in the mouse mitochondrial genome
Pedro Silva-Pinheiro (MRC Mitochondrial Biology Unit, University of Cambridge, United Kingdom)
Click for abstract
MitoKO: A library of base editors for the precise ablation of all protein-coding genes in the mouse mitochondrial genome
Pedro Silva-Pinheiro (1), Christian D. Mutti (1), Lindsey Van Haute (1), Christopher A. Powell (1), PavelA. Nash (1), Keira Turner (1) & Michal Minczuk (1)
1- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
Mitochondria are cellular organelles that have their own genome; mitochondrial DNA (mtDNA) encodes 13 subunits of the oxidative phosphorylation system, all of which are essential for the cell’s energy supply. Mutations in mtDNA can result in mitochondrial diseases, which are a group of incurable conditions affecting approximately 1 in 8,000 humans (1). MtDNA mutations have also been associated with common multifactorial diseases, metabolic disease, heart failure, cancer, neurodegeneration and ageing. Our progress towards understanding mitochondrial biology in health and disease has been hindered by the inability to precisely manipulate or modify mtDNA in mammalian mitochondria in living cells. Consequently, only a few cellular and animal models are available for researching mitochondrial biology. This limitation highlights the need to develop new approaches to manipulate the mitochondrial genome that will enable mtDNA dysfunction to be modelled in vitro and in vivo (2). DNA base editing was previously adapted to operate within mitochondria owing to the development of DddA-derived cytosine base editors (DdCBEs) (3). DdCBEs are based on a modified toxin called DddAtox, which is derived from the bacterium Burkholderia cenocepacia and has been separated into non-toxic halves fused to two adjacent programmable DNA-binding transcription activator-like effector (TALE) proteins that specifically determine the editing site. The resulting protein is targeted to the mitochondrial matrix to catalyse site-specific C to T conversions in mtDNA (or G to A in the opposite strand). We sought to use mitochondrial base editing to generate ‘MitoKO’ — a library of highly specific DdCBEs that can knockout (KO) every protein-coding gene of mouse mtDNA by introducing premature stop codons; our overall intention was to expand the limited repertoire of meaningful in vitro and in vivo models. Our strategy to introduce these premature stops codons was to use DdCBEs to convert TGA codons (encoding Trp (tryptophan)) present early in the mitochondrial gene into TAA codons (encoding STOP) by deaminating the C on the opposite (non-coding) strand (5′ TCA > 5′ TTA). We assembled a library of eight DdCBE designs per open reading frame and screened them in mouse cells for high ontarget editing and low off-target effects. Our approach resulted in a library of 13 specific DdCBEs capable of introducing high levels of truncating mutations into the open reading frames of mouse mtDNA, therefore knocking out each mtDNA-encoded protein-coding gene. We then applied our MitoKO library, generating KO cells in vitro and creating a mouse model with a truncating mtDNA mutation in Atp6 (the gene encoding ATP synthase subunit a) that perturbed the assembly of the ATP synthase complex; this feature is characteristic of some mitochondrial diseases. The MitoKO library should allow the systematic and comprehensive investigation of mtDNA-related pathways by providing an ‘off-the-shelf’ solution for researchers interested in establishing inactivating mutations in mouse mtDNA. It should also help researchers to investigate the fundamental processes that occur in mitochondria and their impact on organismal homeostasis, and to generate clinically meaningful in vivo models of mtDNA dysfunction for drug discovery and pre-clinical investigations.
References:
1. Gorman, G. S. et al. Mitochondrial diseases. Nat. Rev. Dis. Primers 2, 16080 (2016). A review article presenting an overview of mitochondrial diseases.
2. Silva-Pinheiro, P. & Minczuk, M. The potential of mitochondrial genome engineering. Nat. Rev. Genet. 23, 199–214 (2022). A review article that presents the advances of mitochondrial DNA engineering.
3. Mok, B. Y. et al. A bacterial cytidine deaminase toxin enables CRISPR-free mitochondrial base editing. Nature 583, 631–637 (2020). This paper reports the development of the mitochondrial DNA base editor, DdCBE."
15h30 - 16h00 FLASH TALKS (Each is a 5 min oral presentation, complemented with a Poster)
DNM1L mutations cause mitochondrial disfunction and alterations in mitochondrial networks in fibroblasts from patients with neurodevelopmental disorders.
Mariví Cascajo Almenara (Centro Andaluz de Biología del Desarrollo, CIBERER-UPO, Sevilla, Spain)
Click for abstract
DNM1L mutations cause mitochondrial disfunction and alterations in mitochondrial networks in fibroblasts from patients with neurodevelopmental disorders.
Araceli Sama Barroso (1), Ana Sánchez Cuesta (1), José María Urbano Fernández (2), Cristian Domínguez Tristán (4), Alejandro Campoy López (2), Nelmar Valentina Ortiz Cabrera (3), Verónica Cantarín Extremera (3), Carlos Santos Ocaña (1), Mariví Cascajo Almenara (1)
1- Centro Andaluz de Biología del Desarrollo, CABD-CIBERER-UPO, Sevilla.
2- Advanced Light Microscopy and Imaging Unit (ALMIA), CABD-CSIC-UPO, Sevilla.
3- Hospital Infantil Universitario Niño Jesús, Madrid.
4- Universidad de Sevilla
DRP1 is an essential GTPase in mitochondrial cleavage, trafficking, and distribution and is encoded by the Dynamin1-like gene (DNM1L). This protein is produced in the cytosol as a dimer but requires binding to the outer mitochondrial membrane to activate and initiate mitochondrial fission. This recruitment stimulates the protein assembly, which oligomerizes into ring structures, drives membrane constriction, and promotes division following GTP hydrolysis. DRP1 consists of three domains: a middle domain involved in DRP1 oligomerization, a GTPase domain responsible for membrane constriction, and a GED domain that stimulates GTPase activity. Mutations in this gene involve imbalances in mitochondrial function produced by alterations in mitochondrial fission. To date, few patients with mutations in DNM1L have been described. They show a variable and complex phenotype, ranging from hypotonia, cognitive development, developmental delay, and epilepsy to lethal encephalopathy in neonates. The pathogenic variants described in DNM1L are related to the defect in mitochondrial fission resulting from alterations in the DRP1 protein. Due to the various symptoms observed in these patients, it is essential to characterize how DNM1L mutations can alter mitochondrial physiology. In this study, we aim to characterize the structure, morphology, and mitochondrial dynamics in fibroblasts derived from a mother with a speech disorder and pes cavus and her 11-year-old son with global developmental delay, equinus gait, and epilepsy, both with a variant in the DNM1L gene, c.1916G>A; p.Arg639Gln, in heterozygosis, and a child (P131) with a variant in the DNM1L gene: c.1486_1487delinsGA; p.Leu496Asp in heterozygosis de novo.
Impaired mitochondrial response to oxidative stress: differentiating healthy from pathological ageing
Diogo Trigo (Department of Medical Sciences, iBiMED, Universidade de Aveiro, Portugal)
Click for abstract
Impaired mitochondrial response to oxidative stress: differentiating healthy from pathological ageing
Diogo Trigo (1), André Nadais (1), José João Vitória (1), Odete AB da Cruz e Silva (1)
1- Department of Medical Sciences, iBiMED, Universidade de Aveiro
The plastic, dynamic architecture of the mitochondrial network is crucial to maintain energy homeostasis; this role is particularly critical in the brain, due to its high energy demand. However, the physiological properties of mitochondria deteriorate with ageing, which contributes to increase production of reactive oxygen species and macromolecule and organelle dysfunction: mitochondrial loss of efficiency is likely to be both a cause and a consequence of ageing.
Our work has previously showed that the neuroprotective retinoic acid receptor signalling pathway modulates the mitochondria network, but although the correlation between ageing and mitochondria disfunction is well established, a therapeutic protocol promoting mitostasis remains to be established.
This work explores the degradation of mitochondria oxidative stress-response mechanisms with ageing in human cells, addressing the physiological effects on protein aggregations, focused on its role in differentiating between healthy and pathological ageing. Using our novel routine optimization that automatizes analysis of live mitochondria imaging, we observed that impaired response to oxidative stress by aged mitochondria appears tightly related to protein aggregation, and antioxidative agents have a progressive protective effect with age, with cells from old individuals being more susceptible to oxidative stress. Moreover, analysis of the insoluble protein fraction identified novel retinoid-regulated mitochondrial and metabolic proteins, differently expressed with ageing and health condition of donors.
This work shows that the flavonoid catechin prevents the oxidative stress-driven increase in protein aggregation in older cells, supporting the antioxidant properties of flavonoids as a therapeutic strategy for age-related diseases. Cells from old donors are more susceptibility to oxidative stress, in terms of protein aggregation, cell viability, or mitochondria homeostasis. Hence, antioxidative agents, such as flavonoids, can be a strategic therapeutic for several age-related diseases; however, long-term effects on mitochondria and healthy ageing remain unknown, so more studies in this area are necessary. Ongoing work is aiming to use retinoid signalling to regulate mitochondrial response to oxidative stress in pathological conditions to promote homeostasis.
Acknowledgements: This study was funded by the Institute for Biomedicine—iBiMED (UIDB/04501/2020 and UIDP/04501/2020), the Integrated Programme of SR&TD ”pAGE”CENTRO-01-0145-FEDER-000003, PTDC/DTP-PIC/5587/2014, UID/BIM/04501/2013, EXPL/BTM-SAL/0902/2021, FCT (2020.02006.CEECIND, EXPL/BTM-SAL/0902/2021), La Caixa Foundation (CI21-00276), and All Time GABBAs.
Deep mitochondrial genotyping in advanced cellular models of Parkinson's disease indicates altered mitochondrial quality control mechanisms
Valentina Gilmozzi (IBER, University of Lübeck, Bolzano; CIBIO, University of Trento; Italy)
Click for abstract
Deep mitochondrial genotyping in advanced cellular models of Parkinson's disease indicates altered mitochondrial quality control mechanisms
Valentina Gilmozzi (1,2), Martin Lang (1), Irene Pichler (1)
1- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
2- Department of Cellular, Computational and Integrative Biology, (CIBIO) University of Trento, Trento, Italy
Introduction:
Parkinson's disease (PD) is a neurodegenerative movement disorder, characterized by progressive loss of dopaminergic neurons, accompanied by mitochondrial dysfunction. The contribution of mitochondrial DNA (mtDNA) variants to PD pathogenesis has long been debated. Nuclear genes driving inherited forms of PD, such as PRKN, are linked to mtDNA quality control mechanisms. Accordingly, a role of somatic mtDNA variants may play a role in mitochondrial dysfunction in PD. (reviewed in 1)
Materiel & Methods:
mtDNA copy number and major arc deletion were analyzed in whole blood, lymphoblasts, fibroblasts, induced pluripotent stem cells (iPSCs) and iPSC-derived neurons of PD patients, healthy heterozygous PRKN variant carriers, and matched controls by digital droplet-PCR.
D-Loop activity was analyzed in the same samples by realtime-PCR. High coverage whole mtDNA genome sequences were obtained by next generation sequencing.
Results:
An alteration of mtDNA copy number, D-Loop activity, and heteroplasmic variants was observed in iPSCs and -derived neurons as compared to blood cells. The correlation of heteroplasmy loads in iPSCs and neurons between controls and PRKN variant carriers was altered.
Conclusion:
Advanced cellular models are indicated for the study of mitochondrial genotypes and its contribution to the cellular phenotype in PD. Deep mtDNA genotyping implies an altered mitochondrial quality control mechanism in PD-related mutation carriers.
References
1. Lang et al., 2022; Cell Mol Life Sci.; doi:10.1007/s00018-022-04304-3; A genome on shaky ground: exploring the impact of mitochondrial DNA integrity on Parkinson's disease by highlighting the use of cybrid models
The Road So Far and beyond: 28 years of Leber’s Hereditary Optic Neuropathy at LBioMiT - past, present and future
Sara Martins (CIBB, CNC, IIIUC, University of Coimbra, Portugal)
Click for abstract
The Road So Far and beyond: 28 years of Leber’s Hereditary Optic Neuropathy at LBioMiT - past, present and future
Sara Martins, MSc (1,2,3 †), Maria João Santos, MSc (1,2,4 †), Marta Simões (1,2,5), António Friande Pereira (6,7), Cristina Almeida (6,8), Dália Meira (6,9), Gil Resendes (6,10), Joana Tavares Ferreira (6,11,12), João Costa (6,13), Lígia Ribeiro (6,9), Maria Araújo (6,7), Marta Macedo (6,14), Miguel Raimundo (6,15), Olinda Faria (6,16,17), Pedro Fonseca (6,4,15), Sérgio Estrela (6,16,17), João Lemos (15), João Durães (4,15,18), Luísa Diogo (1,4,15,18), Maria Carmo Macário (1,4,15,18), Manuela Grazina, PhD* (1,2,4)
1- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal;
2- Laboratory of Mitochondrial Biomedicine and Theranostics, CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal;
3- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal;
4- FMUC - Faculty of Medicine, University of Coimbra, Coimbra, Portugal;
5- FCTUC – Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal;
6- PT_LHON Taskforce Team;
7- Centro Hospitalar Universitário do Porto, Porto, Portugal;
8- Hospital de Braga, Braga, Portugal;
9- Centro Hospitalar de Vila Nova de Gaia/Espinho, Portugal;
10- Hospital do Divino Espírito Santo de Ponta Delgada, Ponta Delgada, Portugal;
11- Centro Hospitalar Universitário Lisboa Norte - Hospital Santa Maria, Lisboa, Portugal;
12- Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal;
13- Centro Hospitalar de Lisboa Ocidental, Lisboa, Portugal;
14- Hospital Dr. Nélio Mendonça, Funchal, Portugal;
15- CHUC - Centro Hospitalar e Universitário de Coimbra, EPE, Coimbra, Portugal;
16- Faculdade de Medicina da Universidade do Porto, Porto, Portugal;
17- Centro Hospitalar Universitário de São João, Porto;
18- Centro de Referência de Doenças Hereditárias do Metabolismo – CHUC, MetabERN;
† Authors contributed equally to this work
Many cases of optic atrophy, particularly Leber’s Hereditary Optic Neuropathy (LHON), remain without an identified genetic cause[1,2]. The diagnosis of LHON includes screening for mitochondrial DNA (mtDNA) variants, namely TOP3: m.3460G>A, m.11778G>A, and m.14484T>C (TOP3)[4]. In the last 28 years, 141 cases were assigned to our laboratory. Recent technological advances bring additional challenges and identification of novel variants. Biochemical/functional studies are crucial for their validation[3]. GenEye24[4] ensures TOP3 screening within 24h. The results of LHON screening at LBioMiT are presented. Genetic screening was performed by NGS and bioinformatic analyses[3]. Biochemical evaluation of OXPHOS complexes activities was done by double wavelength spectrophotometry[5]. The GenEye24 relies on real-time PCR with High-Resolution Melting[4]. Whole mtDNA screening grouped: 124 patients with unidentified mtDNA cause (88%); 17 patients with mtDNA variants (12%) – 59% m.11778G>A; 17% m.14484T>C; 24% m.3460G>A. A hundred and eight samples were genotyped with GenEye24 testing: 64 wild-type, 36 m.11778G>A, 6 m.14484T>C and 2 m.3460G>A, with high confidence values (Mode=100%). Sensitivity and specificity were both 1. Results highlight the need for a complete genetic screening. The GenEye24 is a fast, robust, simple and cost-effective alternative to TOP3 screening. Fast results allow for timely treatment and visual function rescue. The project “Translational Epidemiological, Bigenomics and Functional Research in Optic Atrophies” (approved by ethical committees in 11 hospitals) allows centralization of the LHON bigenomic study in Portugal.
Support:
This work was financed by the European Regional Development Fund (ERDF), through the COMPETE 2020 - Operational Programme for Competitiveness and Internationalisation and Portuguese national funds via FCT – Fundação para a Ciência e a Tecnologia, under project[s] POCI-01-0145-FEDER-007440; CENTRO-01-0145-FEDER-000012-N2323, CENTRO-07-ST24-FEDER-002002/6/, UID/NEU/04539/2013, Pest-C/SAU/LA0001/2013-2014, FCTSFRH/ BD/86622/2012 and UIDB/04539/2020, UIDP/04539/2020 and LA/P/0058/2020. The LBioMiT was financed by Santhera Pharmaceuticals, allowing the implementation of the project “Providing free of charge complete genetic tests to Portuguese patients with a clinical and instrumental diagnosis of Optic Nerve Atrophy” (PI M Grazina)
References:
1. Grazina et al. Eur J PaediatrNeurol. 2007 Mar;11(2):115-8.
2. Carelliet al. Hum Mol Genet. 2017 Oct;26(R2):R139-R150.
3. Bacalhau et al. NeuromusculDisord. 2018 Apr;28(4):350-360.
4. Martins et al. Mitochondrion 2023 Feb. DOI:10.1016/j.mito.2023.01.006
5. Grazina MM. Methods Mol Biol. 2012;837:73-91.
16h00-17h00
Coffee-Break + Posters
Foyer
17h00-18h30
Session IV
Salão Nobre
Chairs
Andrea Irazoki, Paulo J. Oliveira
17h00-17h30
Mitochondria quality control and neurodegeneration
Vanessa Morais (iMM - Institute of Molecular Medicine, Portugal)
Click for Abstract
Mitochondria are organelles known as the “powerhouses” of the cells, as they are responsible for producing 90% of the energy necessary to maintain and support cell survival. When mitochondria fail to produce enough energy to sustain cell life, degeneration of the cells, the tissue and ultimately the organ occurs.
As the brain is a high energy demanding organ, our overarching goal is to clarify the intimate crosstalk between the host cell – the neuron – and the powerhouse organelle – the mitochondria. Mitochondria at the synapse have a pivotal role in neurotransmitter release, but almost nothing is known about synaptic mitochondria composition or specific functions. Synaptic mitochondria compared to mitochondria in other cells, need to cope with increased calcium load, more oxidative stress, and high energy demands that sustain neurotransmitter release and synaptic plasticity. At present, we have deciphered the intrinsic properties of synaptic mitochondria. For this, we have assessed the protein fingerprint, the metabolic profile and fuel preference of synaptic mitochondria. Ultimately, we intend to unveil how disruption of these synapse-specific mechanisms are relevant for a healthy brain, and to scrutinize their relevance in neurodegenerative disorders.
17h30 - 17h45
GEMIN5 and CoQ10, a link between neurodevelopment and mitochondrial metabolism
Carlos Santos Ocaña (Universidad Pablo de Olavide, Sevilla, Spain)
Click for Abstract
GEMIN5 and CoQ10, a link between neurodevelopment and mitochondrial metabolism
María Victoria Cascajo Almenara (1,3), Ana María Sánchez Cuesta (1,3), Daniel M. Fernández Ayala (1,3), Elena García Díaz (1), Rafael Artuch (2,4), and Carlos Santos Ocaña (1,3)
1-Universidad Pablo de Olavide, Sevilla, Spain
2-Hospital San Joan de Deu, Barcelona, España
3-CIBERER U729,
4-CIBERER U703
Introduction
In 2006 a case of CoQ10 deficiency with cerebellar ataxia that improved after treatment with CoQ10 was published (1). However, a definitive molecular diagnosis could not be obtained. Recently, exome sequencing has shown that the patient carries a biallelic mutation in GEMIN5, a component of the SMN (Survival Motor Neuron) complex involved in transcriptional regulation (2). Searching for similar patients has allowed us to find a second patient already included in an extensive study of patients with cerebellar atrophy and delayed motor development (3) and a third patient who died a few months after birth.
Results
Patient fibroblasts show CoQ10 deficiency, decreased mitochondrial mass, and mitochondrial dysfunction manifested by low oxygen consumption, decreased activity of complexes II, III, and II+III of the respiratory chain, mtDNA depletion, and decreased expression of COQ proteins. Incorporating the 13C-labeled 4-HB precursor made it possible to determine a decrease in CoQ10 synthesis by LC-MS. The transcriptomic analysis in fibroblasts shows repression of mitochondrial biogenesis, fatty acid oxidation, Krebs cycle, OXPHOS activity, and skeletal muscle development. Transcriptional repression specifically affects genes encoding proteins required for CoQ10 synthesis, PPTC7 phosphatase, and transcription factors such as PGC-1α.
Functional validation of GEMIN5 deficiency was addressed in a silencing model of its orthologous gene, Rigor Mortis (rig), in D. melanogaster. Silencing increases mortality, increases the number of non-motile flies, decreases climbing ability, and decreases CoQ levels (CoQ8, CoQ9, and CoQ10). Analysis of larval development shows that silencing reduces the number of adult flies hatched.
In order to address the use of CoQ10 as a therapeutic strategy, CoQ10 was administered to fibroblasts from GEMIN5 patients achieving a recovery of total CoQ10 levels and oxygen consumption. In the fly model, CoQ10 supplementation increases only CoQ10 levels but no CoQ8 and CoQ9. However, it increases survival and restores the number of adult flies hatched during larval development to control levels.
Conclusions
Patient-derived fibroblasts show impaired mitochondrial function with a CoQ10 deficiency that can be termed secondary by current clinical criteria (no COQ genes are affected) but show some characteristics of a primary CoQ10 deficiency. In the D. melanogaster model, GEMIN5 silencing recapitulates the defects observed in patients, and as in patient fibroblasts, CoQ10 supplementation significantly restores these defects. Therefore, CoQ10 may constitute a suitable therapeutic strategy for treating neuro-disorders caused by defects in the GEMIN5 gene.
References
1. Artuch R, et al (2006). J. Neurol. Sci. 246: 153–158. 10.1016/j.jns.2006.01.021
2. Martinez-Salas E, et al (2020). Int. Journal Mol, Sci. 21: 3868. 10.3390/ijms21113868
3. Kour S, et al (2021). Nat Commun 12: 2558, 10.1038/s41467-021-22627-w
17h45 - 18h00
Alterations in the contacts between endoplasmic reticulum and mitochondria in Bipolar disorder
Ana Catarina Pereira (CNC, FMUC, CACC, University of Coimbra, Portugal)
Click for Abstract
Alterations in the contacts between endoplasmic reticulum and mitochondria in Bipolar disorder
Ana Catarina Pereira (1,2,3), Ana Patrícia Marques (1,3,4), Rosa Resende (1,3,4), Laura Serrano-Cuñarro (1), Mariana Batista (5), António Macedo (2,3,6,7), Cláudia Pais (8), Joana B. Melo (2,3,8,9), Nuno Madeira (2,3,6,7), Maria Teresa Cruz (1,3,10), Cláudia Cavadas (1,3,10), Cláudia Pereira (1,2,3)
1-CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal;
2-Faculty of Medicine, University of Coimbra, Coimbra, Portugal;
3-CACC - Clinical Academic Center of Coimbra, Coimbra, Portugal;
4-IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal;
5-CHUC - Centro Hospitalar e Universitário de Coimbra, Department of Dermatology, Coimbra, Portugal;
6-CHUC - Centro Hospitalar e Universitário de Coimbra, Department of Psychiatry Coimbra, Coimbra, Portugal;
7-CIBIT - Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal;
8-Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, Portugal;
9-iCBR - Coimbra Institute for Clinical and Biomedical Research, CIMAGO - Center of Investigation in Environment, Genetics and Oncobiology, Coimbra, Portugal;
10-Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
Mitochondria-Associated Membranes (MAMs), which are the contact sites between the endoplasmic reticulum (ER) and mitochondria, are dynamic platforms that regulate several key processes, particularly mitochondrial bioenergetics and dynamics, as well as stress responses, due to the transfer of Ca2+ and lipids between both organelles. MAMs are modulated according to the cellular needs, and the impairment of these stress response structures has been linked to pathological conditions closely associated with mitochondrial dysfunction and ER stress, among other deleterious events. Since these molecular alterations have been closely associated with the structural brain changes and cognitive deficits reported in Bipolar disorder (BD), this study aimed to investigate the role of MAMs in BD physiopathology.
Dermal fibroblasts from BD patients versus healthy matched controls were used as an in vitro model of this mental illness. The ER-mitochondria coupling was assessed by confocal microscopy and transmission electron microscopy. Concomitantly, MAMs functional parameters such as Ca2+ fluxes, reactive oxygen species (ROS) production, and lipid droplets formation, were determined using the Fluo-4/Rhod-2, Mitopy/CellROX and LipidTOX fluorescent probes, respectively. Mitochondrial bioenergetics were evaluated by the Seahorse assay. Protein levels of ER stress-induced Unfolded Protein Response (UPR) markers were assessed by Western Blot, and autophagy was investigated by measuring the immunoreactivity of the autophagy-associated protein LC3B.
In BD patients-derived fibroblasts, it was observed an increase in the number of close ER-mitochondria contacts sites, or MAMs, which was shown to be correlated with functional alterations, such as deregulated ER-to-mitochondria Ca2+ transfer, enhanced ROS production, ER-to-mitochondria lipids transfer and formation of Lipid droplets, ER stress induction, as well as perturbation of mitochondrial metabolism and autophagy.
This study shows that the impaired communication between the ER and mitochondria at MAMs is implicated in BD physiopathology and might impact several crucial cellular and molecular events leading to the clinical manifestations of the pathology.
Acknowledgements: European Regional Development Fund (ERDF), through the Centro 2020 Regional Operational Programme under project CENTRO-01-0145-FEDER-000012 (HealthyAging2020) and through the COMPETE 2020 - Operational Programme for Competitiveness and Internationalisation and Portuguese national funds via FCT – Fundação para a Ciência e a Tecnologia, under projects POCI-01-0145-FEDER-028214 (MAM4BD) and POCI-01-0145-FEDER-029369 and UIDB/04539/2020 and UIDP/04539/2020. PhD fellowship from FCT (SFRH/BD/148653/2019) to Ana Catarina Pereira. European Social Fund (Post-Doctoral Researcher Contract SFRH/BPD/101028/2014 to Rosa Resende).
18h00 - 18h15
The emerging role of coenzyme A and protein CoAlation in redox regulation
Ivan Gout (University College London, United Kingdom)
Click for Abstract
Coenzyme A (CoA) is a key metabolic cofactor in all living cells. CoA and its thioester derivatives (acetyl-CoA, malonyl-CoA, HMG-CoA etc.) participate in diverse anabolic and catabolic pathways, allosteric interactions, biosynthesis of neurotransmitters and the regulation of gene expression. Dysregulation of CoA/CoA derivatives biosynthesis and homeostasis has been associated with various human pathologies, including cancer and neurodegeneration and metabolic disorders.
We have recently discovered a novel mode of redox regulation, involving covalent modification of cellular proteins by CoA (protein CoAlation) in cellular response to oxidative or metabolic stress. To discover and study this novel post-translational modification, we have developed several novel reagents and methodologies, including: (a) anti-CoA mAb, which specifically recognize CoA in ELISA, WB, IP and IHC (no anti-CoA antibodies are commercially available); (b) a robust mass spectrometry-based methodology for the identification of CoAlated proteins; and (c) efficient in vitro CoAlation and deCoAlation assays. Cell-based and animal models have been employed to demonstrate that protein CoAlation is a reversible and widespread post-translational modification induced by oxidizing agents and metabolic stress in prokaryotic and eukaryotic cells. To date, we have identified using the developed methodology more than 2100 proteins, which are CoAlated under various experimental conditions in cell and tissues. We showed that protein CoAlation modulates the activity and subcellular localization of modified proteins. It can also protect oxidized cysteine residues from overoxidation and induce significant conformational changes. We have employed biochemical, biophysical, crystallographic and cellular approaches to study the mode of CoA binding to a panel of selected metabolic enzymes and signalling proteins. Over the last 5 years we have firmly established ourselves as a world-leading laboratory on protein CoAlation and the antioxidant function of CoA in health and pathologies associated with oxidative stress, including cancer and neurodegeneration.
Recent progress on molecular dissection of the CoAlation/deCoAlation cycle and investigating the antioxidant function of CoA in pathologies associated with oxidative stress will be presented.
18h15 - 18h30 FLASH TALKS (Each is a 5 min oral presentation, complemented with a Poster)
Intercellular mitochondrial transfer via tunnelling nanotubes: a redox regulated process?
Emily Glover (University of Bristol, United Kingdom)
Click for Abstract
Intercellular mitochondrial transfer via tunnelling nanotubes: a redox regulated process?
Emily Glover, Hope Needs, Ian Collinson
Tunnelling nanotubes (TNTs) are actin-based cell protrusions, first described in 2004 by Rustom et al. as ‘open-ended channels mediating membrane continuity between connected cells’ [1]. This continuity gives TNTs the ability to transfer cargo between connected cells, presenting a means of direct cell-to-cell communication. TNT-mediated mitochondrial transfer has been documented in a range of cell types in cell culture, but a mechanistic understanding of TNT induction and TNT-mediated mitochondrial transfer is lacking [2].
TNTs are typically observed during situations of cellular stress [3] and accumulating evidence suggests that TNT-mediated intercellular mitochondrial transfer is regulated by reactive oxygen species [4]. Indeed, hydrogen peroxide is known to influence F-actin dynamics both directly and indirectly [5,6]. Recent work in the Collinson lab has shown that chronic mitochondrial import failure can be rescued by intercellular mitochondrial transfer through TNTs [7]. We hypothesise that the mechanism for this TNT-mediated rescue is dependent on hydrogen peroxide. At the symposium I would like to present my findings and discuss my plans to further interrogate this phenomenon.
Acknowledgements: Hope Needs, Ian Collinson.
1. Rustom A, Saffrich R, Markovic I, Walther P, Gerdes HH: Nanotubular Highways for Intercellular Organelle Transport. Science 2004, 303:1007–1010.
2. Ljubojevic N, Henderson JM, Zurzolo C: The Ways of Actin: Why Tunneling Nanotubes Are Unique Cell Protrusions. Trends Cell Biol 2021, 31:130–142.
3. Wang X, Gerdes HH: Transfer of mitochondria via tunneling nanotubes rescues apoptotic PC12 cells. Cell Death Differ 2015, 22:1181–1191.
4. Rustom A: The missing link: does tunnelling nanotube-based supercellularity provide a new understanding of chronic and lifestyle diseases? rsob 2016, 6.
5. DalleDonne I, Milzani A, Colombo R: H2O2-treated actin: assembly and polymer interactions with cross-linking proteins. Biophys J 1995, 69:2710–2719.
6. Nimnual AS, Taylor LJ, Bar-Sagi D: Redox-dependent downregulation of Rho by Rac. Nat Cell Biol 2003, 5:236–241.
7. Needs HI, Pereira GC, Glover E, Witt A, Hübner W, Dodding MP, Henley JM, Collinson I: Intercellular Mitochondrial Transfer as a Rescue Mechanism in Response to Protein Import Failure. bioRxiv 2022, doi:10.1101/2022.11.30.518494.
MitoDREADD-Gs: a new tool to acutely increase mitochondrial activity and rescue cognitive alterations
Luigi Bellocchio (INSERM, U1215 NeuroCentre Magendie, University of Bordeaux, France)
Click for Abstract
MitoDREADD-Gs: a new tool to acutely increase mitochondrial activity and rescue cognitive alterations
Antonio C. Pagano Zottola (1,2,*), Rebeca Martin-Jimenez (3,4,*), Gianluca Lavanco (1,2,*), Geneviève Hamel-Côté (3,4,*), Carla Ramon-Duaso (5), Yamuna Mariani (1,2), Stephanie Jean (3,4), Mehtab Khan (3,4), Sandra Beriain (1,2), Astrid Cannich (1,2), Laura Vidal-Palencia (5), Francisca Julio-Kalajzić (1,2), Doriane Gisquet (1,2), Arnau Busquets-Garcia 5, Giovani Marsicano (1,2,#), Etienne Hebert-Chatelain (3,4,#), Luigi Bellochio (1,2,#)
1- INSERM, U1215 NeuroCentre Magendie, Endocannabinoids and Neuroadaptation, Bordeaux, France
2- University of Bordeaux
3- Canada Research Chair in Mitochondrial Signaling and Physiopathology
4- Department of biology, University of Moncton, Canada
5- IMIM-Hospital del Mar Medical Research Institute, PRBB, Barcelona, Spain.
* Equally contributing
# Equally supervising
Most brain mitochondrial defects lead to cognitive impairment or neurodegenerative diseases. However, due to the lack of suitable tools, no direct link between acute mitochondrial activity and higher brain functions has been established so far. Heterotrimeric guanine nucleotide-binding (G) proteins are key players in brain metabolism and higher functions. Since G proteins can be found within mitochondria, we hypothesized that stimulation of specific G protein signaling within the organelle could modulate brain mitochondrial activity and possibly rescue behavioral defects associated to brain metabolic disorders. We developed mitoDREADD-Gs, a Galphas-linked recombinant designer receptor exclusively activated by designer drugs targeted to mitochondria, which can acutely increase mitochondrial metabolism in different types of cells both in vitro and ex vivo. Strikingly, in vivo activation of mitoDREADD-Gs expressed in specific brain circuits abolished cognitive impairments linked to mitochondrial alterations, including cannabinoid-induced amnesia and other memory impairment models. Our data show that mitoDREADD-Gs is a reliable tool to acutely increase mitochondrial activity. This will not only benefit our understanding of how mitochondria are involved in biological functions, but it will also provide new potential therapeutic concepts for the treatment of brain diseases associated to impaired cell metabolism.
18h30
Porto Wine
Foyer
Saturday, May 13, 2023
09:00 - 10h30
Session V
Salão Nobre
Chairs
Vanessa Morais, Mike Murphy
9h00-9h30
Mitochondrial dynamics as a hub in the control of muscle inflammation
Andrea Irazoki (Dep. Biomedical Sciences, University of Copenhagen, Denmark)
Click for Abstract
Mitochondrial dynamics as a hub in the control of muscle inflammation
Andrea Irazoki (1,2,3), Isabel Gordaliza-Alaguero (1, 2, 3), Emma Frank (4), Nikolaos Nikiforos Giakoumakis (1), Jordi Seco (1), Manuel Palacín (1, 2, 5), Anna Gumà (2,3,6), Lykke Sylow (4,7), David Sebastián (1,2,3*) and Antonio Zorzano (1,2,3*)
1-Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10-12, 08028 Barcelona, Spain. 2-Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain. 3-Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) Instituto de Salud Carlos III, Spain. 4-Department of Biomedical Sciences (BMI), University of Copenhagen, Denmark, 5-CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain. 6-Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain. 7-Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark. *Corresponding and supervising authors.
Some forms of mitochondrial dysfunction induce sterile inflammation through mitochondrial DNA (mtDNA) recognition by intracellular DNA sensors. However, the involvement of mitochondrial dynamics mitigating such processes and their impact on muscle fitness remain unaddressed. Here we report that opposite mitochondrial morphologies induce distinct inflammatory signatures, caused by differential activation of DNA sensors TLR9 or cGAS. In the context of mitochondrial fragmentation, we demonstrate that mitochondria-endosome contacts mediated by the endosomal protein RAB5C are required in TLR9 activation in cells. Skeletal muscle mitochondrial fragmentation promotes TLR9-dependent inflammation, muscle atrophy, reduced physical performance and enhanced IL6 response to exercise, which improved upon chronic anti-inflammatory treatment. Taken together, our data demonstrate that mitochondrial dynamics is key in preventing sterile inflammatory responses, which precede the development of muscle atrophy and impaired physical performance. Thus, we propose the targeting of mitochondrial dynamics as an approach to treating disorders characterized by chronic inflammation and mitochondrial dysfunction.
9h30 - 9h45
Non-canonical nitrogen metabolism in Fumarate Hidratase-deficient cells.
Marc Segarra-Mondejar (CECAD Research Centre, University of Cologne, Germany)
Click for Abstract
Non-canonical nitrogen metabolism in Fumarate Hidratase-deficient cells.
Marc Segarra-Mondejar (1), Ming Yang (1), Agnieszka Sokol (1), Christian Frezza (1)
1- CECAD Research Centre, University of Cologne, Cologne, Germany
Fumarate hydratase (FH) is the enzyme in the Tricarboxylic Acid (TCA) Cycle that catalyses the reversible conversion of succinate to fumarate. Germline mutations of FH has been shown to predispose to leiomyomatosis and renal cancer (HLRCC), a cancer syndrome characterized by the development of benign skin and uterine lesions and of an aggressive form of type II papillary renal cell cancer (pRCC2).
We have shown that FH-deficient cells depend on glutamine as the main source of carbons for the TCA cycle and for the synthesis a glutathione, via glutamate, to compensate for the increased oxidative stress that we reported. This reaction is usually catalysed by glutaminase (GLS), which release glutamine amide group in the form of NH4+, a toxic metabolite. However, how FH-deficient cells overcome ammonia toxicity is currently unknown.
Here we found that FH-deficient mouse cells depend on de novo nucleotide and asparagine synthesis pathways to maintain high rates of glutamine catabolism minimising ammonia production. Of note, despite exhibiting a reduction in total pyrimidine levels, FH-deficient cells exhibit a significant increase in early intermediate in pyrimidine synthesis, including carbamoyl-aspartate, and a higher incorporation of glutamine-derived nitrogen into GMP and IMP, and into asparagine. Since all these pathways use the amide-group of glutamine as a source of nitrogen, we conclude that FH-deficient cells rely on transamidases as an alternative mechanism of glutamine deamination. Our results shed some light into FH-deficient tumour metabolism and indicate transamidases inhibitors as a possible therapy against one of the most aggressive forms of kidney cancer.
9h45 - 10h00
AntiOxCIN4 protects against brain and skeletal muscle oxidative/nitrosative stress in the amyotrophic lateral sclerosis SOD1G93A mouse
Ana I. Duarte (CNC, CIBB, IIUC, University of Coimbra, Portugal)
Click for Abstract
AntiOxCIN4 protects against brain and skeletal muscle oxidative/nitrosative stress in the amyotrophic lateral sclerosis SOD1G93A mouse
Débora Mena (1,2,3), Fernando Cagide (4), Sofia Benfeito (4), Pedro Soares (4), Luís Grilo (1,2,3), Daniela F. Silva (1), Paulo Pinheiro (1), Elisabete Ferreiro (1,2), José Teixeira (1,2), Filomena Silva (1), Fernanda Borges (4), Paulo J. Oliveira (1,2,3), Ana I. Duarte (1,2,3*)
1- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal;
2- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal;
3- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal;
4- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal.
Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disease. It is characterized by progressive motor neuron loss, muscle weakness, atrophy and paralysis, ultimately leading to death. Although ALS pathophysiology remains poorly understood, evidence suggests a pivotal role for mitochondrial dysfunction and oxidative stress. The currently available treatments for ALS are only symptomatic and with limited efficacy. Hence, the improvement of mitochondrial function and/or antioxidant capacity may constitute potential therapeutic approaches to delay ALS progression. We hypothesized that the mitochondria-targeted antioxidant AntiOxCIN4 can counteract brain and skeletal muscle oxidative/nitrosative stress in SOD1G93A ALS mice.
Early adult SOD1G93A ALS mice were subcutaneously injected with AntiOxCIN4 (0.1 mg/Kg/day), for 2 months. We obtained brain cortical and skeletal muscle homogenates and used colorimetry/fluorimetry-based methods to assess the effect of AntiOxCIN4 in oxidative/nitrosative stress markers nitrites and hydroperoxides, as well for measuring the activities of the antioxidant enzymes superoxide dismutase (total SOD, SOD-2), and glutathione peroxidase (GPx).
We observed that AntiOxCIN4 upregulates brain GPx activity (P=0.07), decreases skeletal muscle hydroperoxides (P=0.009), and brain and skeletal muscle nitrites levels (P=0.002, P0.001, respectively) in ALS mice. This was accompanied by a slight increase in brain total SOD and SOD-2 activities, as well as by a reduction in total SOD and SOD-2 activities in skeletal muscle (P=0.07, P=0.08, respectively) of AntiOxCIN4-treated ALS mice.
Our results suggest that the peripheral administration of AntiOxCIN4 may counteract ALS brain and skeletal muscle oxidative/nitrosative stress. However, further studies are needed to determine whether this protection delays ALS progression.
This work was funded by European Regional Development Fund: Centro2020 Operational Programme (POCI-01-0145-FEDER-029391 (Mito4ALS)), COMPETE 2020 and FCT- Fundação para a Ciência e a Tecnologia (POCI-01-0145-FEDER-029391, PTDC/MED-FAR/29391/2017, EXPL/BTM-TEC/1407/2021, UIDB/04539/2020, UIDP/04539/2020, LA/P/0058/2020, UIDB/00081/2020); European Social Fund: 2021.04707.BD and Mito4ALS-PTDC/MED-FAR/29391/2017 of D.M.; FCT and DL57/2016 of F.C.; DL57/2016 SFRH/BPD/84473/2012 of A.I.D.; FCT/P2020/COMPETE PTDC/MED-QUI/29164/2017 and PT-OPENSCREEN-NORTE-01-0145-FEDER-085468 of S.B.; EU Horizon2020 R&I Program/Marie Sklodowska-Curie grant 895144 of P.S.; FCT Contract 2020.01560.CEECIND of J.T.; SFRH/BD/5539/2020 of L.G.
10h00 - 10h30 FLASH TALKS (Each is a 5 min oral presentation, complemented with a Poster)
The unique bioenergetic fingerprint of synaptic mitochondria
Andreia Faria-Pereira (iMM - Institute of Molecular Medicine, Portugal)
Click for Abstract
The unique bioenergetic fingerprint of synaptic mitochondria
Andreia Faria-Pereira (1), Marco Spinazzi (2), Katleen Craessaerts (2), Jeffrey Savas (3), Bart De Strooper (2), Vanessa A. Morais (1, 2)
1- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1990-375 Lisboa, Portugal
2- VIB Center for the Biology of Disease, 3000 Leuven, Belgium; Center for Human Genetics, KU Leuven, 3000 Leuven, Belgium
3- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
The brain is one of the most energy-demanding organs. Most of the energy used in the brain is required for synaptic transmission. Being synapses one of the highest energy-consuming brain structures, as well as one of the most molecularly dynamic sites in the brain - within milliseconds they can change from a resting to a stimulated state, it is expected that mitochondria at synapses – synaptic mitochondria - are adapted to the synaptic environment and present distinct properties from other brain mitochondria - non-synaptic mitochondria.
Indeed, differences at the level of morphology, lipidomics, enzymology and damage-susceptibility have been found between synaptic and non-synaptic mitochondria. Nonetheless, the functional consequences of these differences and more specifically whether synaptic mitochondria have specialized bioenergetic properties adapted to the synaptic environment remains unclear.
By dissecting the bioenergetic profile of brain mitochondria, our results unveiled that synaptic mitochondria have a higher bioenergetic flexibility to respond to different respiratory stimulus. Also, we observed a higher enzymatic activity of Complex IV and Complex V, which are accompanied by the presence of an energetically-favourable molecular arrangement of Complex V. Moreover, synaptic mitochondria present a decreased enzymatic activity of individual Complex I, accompanied by a putative N-module expression and functional signature. However, the enzymatic activity of the supercomplex I and III, as well as Complex I integration in supercomplexes were enhanced in synaptic mitochondria.
We hypothesised that this dicotomic Complex I pattern is a functional bioenergetic signature of synaptic mitochondria that might constitute a protective yet efficient mechanism so that synaptic mitochondria in resting conditions are still able to provide ATP for basal synaptic activity but keeping low ROS damage derived from individual Complex I activity.
These distinct bioenergetics and metabolic features may constitute the necessary flexibility for synaptic mitochondria to adapt to the bioenergetics demanding environment at synapses.
Since neurodegenerative disorders’ early hallmarks consist of energy deficits and loss of synapses, our results will set a groundwork to study the underlying causes of synaptic energy deficits in early asymptomatic stages of neurodegenerative disorders.
A new view on the role of mito-ribosomal fidelity on human disease: structural analysis of deafness-related mtDNA variants mapping to mitochondrial rRNA genes
Antón Vila-Sanjurjo (GIBE, CICA, Universidade da Coruña, Spain)
Click for Abstract
A new view on the role of mito-ribosomal fidelity on human disease: structural analysis of deafness-related mtDNA variants mapping to mitochondrial rRNA genes
Antón Vila-Sanjurjo (1), Natalia Mallo (1), John Atkins (2); Joanna L. Elson (3,4), Paul M. Smith (5), Emma L. Blakely (6,7), & Robert W. Taylor (6,7)
1- Grupo GIBE. Departamento de Bioloxía e Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña (UDC). Campus Zapateira, s/n, 15071, A Coruña, Spain. Electronic address: anton.vila@udc.es.
2- Schools of Biochemistry and Microbiology, University College Cork. T12 XF62 Ireland.
3- The Bioscience Institute, Newcastle University, Newcastle upon Tyne, NE1 3BZ, United Kingdom
4- Human Metabolomics, North-West University, Potchefstroom, South Africa.
5- Department of Paediatrics, Raigmore Hospital, Inverness, Scotland, UK
6- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
7- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE1 4LP, UK
Exploring the impact of metabolic syndrome on brain stiffness and metabolism: Insights from in vivo and in vitro experiments
Heloísa Gerardo (CNC, CIBB, University of Coimbra, Portugal)
Click for Abstract
Exploring the impact of metabolic syndrome on brain stiffness and metabolism: Insights from in vivo and in vitro experiments
Heloísa Gerardo (1,2), Ricardo Amorim (1,3), Inês C.M. Simões (4), Cláudia Cavadas (1), Célia Aveleira (5), Mariusz R. Wieckowski (4), Paulo J. Oliveira (1), Mário Grãos (1) and José Teixeira (1)
1- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
2- Doctoral Programme in Pharmaceutical Sciences, Faculty of Pharmacy, University of Coimbra, 3004-504 Coimbra, Portugal
3- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007, Portugal
4- Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
5- MIA – Multidisciplinary Institute of Ageing, University of Coimbra, 3000-370 Coimbra, Portugal
Metabolic syndrome (MetS) affects a quarter of the population worldwide, representing a major public health concern. MetS is a clustering of several disorders including dyslipidemia, obesity and hyperglycemia/insulin resistance, conditions all linked to an increased risk of neurodegenerative diseases like Alzheimer's and dementia[1]. Although the mechanisms behind these diseases are not fully understood, impairment in brain tissue mechanics have been observed. This comprises specifically a decrease in brain stiffness, which can affect metabolic pathways[2]. Although impairments in mechanotransduction is likely to induced metabolism dysfunction and vice-versa, the role of these complex processes as upstream initiators of disease pathology and progression is still poorly understood[3].
Hence, we performed a study on mice fed with a standard diet (SD) versus Western diet (WD) that induces metabolic syndrome to explore the impact of this condition on brain stiffness and metabolism. We also investigated in vitro a possible cause-effect relationship between brain stiffness alteration and these metabolic processes, we cultured mouse hippocampal neuronal cell line on hydrogels resembling brain tissue softening in dementia[4] (physiological (~6.5-7.5kPa) or dementia (2.5/2.0kPa) resembling brain stiffness). We found that WD-fed mice had decreased levels of proteins associated with brain stiffness (α-SMA, cofilin 30% and CTGF 30%), but no changes in mitochondrial markers (TOM20, VDAC, ATP5). However, we observed an increase in markers related to glucose metabolism (GLUT1 200%, HKII 30%) and mitochondrial beta-oxidation (CPT1 125%). In vitro, culturing cells on hydrogels with stiffness resembling physiological (6.5kPa) or dementia (2.5kPa) impacted cells’ mechanotransduction signals (p-cofilin/cofilin ratio 65%, YAP nuclear to cytoplasmic ratio 25%), as expected. Interestingly, there is an overall decay of protein levels associated with mitochondrial, glycolytic or fatty acid oxidation metabolism (Complex IV 25%, HK 80%, HADHA 55%) when cells were cultured on 2.5kPa substrate compared to 6.5kPa.
Our results suggest that primary alterations in brain lipid metabolism may lead to subsequent changes in brain tissue mechanics in mice with metabolic syndrome induced by a Western diet.
Acknowledgements: This work was financed by the European Regional Development Fund (ERDF), through the Centro 2020 Regional Operational Programme and through the COMPETE 2020 - Operational Programme for Competitiveness and Internationalisation and Portuguese national funds via FCT, under project[s]: EXPL/BIA-BQM/1361/2021, MitoBOOST V2.0 IT137-22-151, CENTRO-01-0246-FEDER-000010 (Multidisciplinary Institute of Ageing in Coimbra), UIDP/04539/2020 and LA/P/0058/2020. J. Teixeira (2020.01560.CEECIND) and H. Gerardo (SFRH/BD/147316/2019) acknowledges FCT, I.P. for the research contract.
1- https://doi.org/10.3390/nu14020333
2- PMID: 28993232
3- PMID: 35177291
4- PMID: 22326238
10h30 - 11h15
Coffee-Break + Posters
Foyer
11h15 - 12h15
Session VI
Salão Nobre
Chairs
Cristina Rego, Jorge MA Oliveira
11h15 - 11h30 Oral communication (10-15 min)
Mitochondrial replication at the periphery of neurons requires eEF1A1 and local translation of nuclear-encoded proteins
Carlos Ramos (iMM - Institute of Molecular Medicine, Portugal)
Click for Abstract
Mitochondrial replication at the periphery of neurons requires eEF1A1 and local translation of nuclear-encoded proteins
Carlos Cardanho-Ramos (1), Rúben A. Simões (1), Andreia Faria-Pereira (1), Vanessa A. Morais (1)
1- Instituto de Medicina Molecular - João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
Neurons rely on mitochondria for ATP production and Ca2+ homeostasis, particularly at the synapse. It is assumed that mitochondria are generated in the cell body and travel to the synapse to exert their functions. However, considering the rate of mitochondrial transport in neurons, the time it would take for a single mitochondrion to travel from the cell body to the synapse exceeds the half-life of most mitochondrial proteins. Deficits in mitochondrial replication have been linked to several neurodegenerative diseases, such as Parkinson’s and Huntington’s disease, however whether mitochondria can replicate locally in the periphery of neurons is still unknown.
We developed a technique to assess mitochondrial replication in mouse primary neuron cultures using EdU-labelling, a thymidine analogue that is incorporated into mtDNA upon replication. Most EdU staining was observed in the cell body, but staining was also present to a lower extent in the periphery. Using microfluidic devices, where axons can be isolated from the cell body, we were able to add EdU only to the axonal side, without interfering with the cell body. In these conditions, EdU staining was only present in axons, confirming that mitochondrial replication in neurons can occur away from the cell body. We hypothesized that mRNA and local translation must be at place in the periphery of neurons in order to provide all the necessary machinery for mitochondria to replicate. To test this, we assessed mitochondrial replication upon inhibition of both nuclear-encoded and mitochondrial-encoded protein translation. Mitochondrial replication in neurons decreased when nuclear-encoded protein translation was inhibited. However, no differences were observed upon inhibition of mitochondrial-encoded protein translation.
Taking advantage of a proteomic screen comparing synaptic with non-synaptic mitochondria, two candidate proteins related with protein translation were found upregulated in the synaptic fraction – eEFF1a1, involved in nuclear-encoded protein translation; and TUFM, involved in mitochondrial-encoded protein translation. We performed loss and gain of function assays with our candidate proteins and assessed their impact on mitochondrial replication. When eEF1A1 was downregulated, we observed a decrease in mitochondrial replication in the periphery of neurons. This effect was rescued by re-introducing eEF1A1. Regarding TUFM, no differences were observed.
Our results confirm that mitochondrial replication can occur in the periphery of neurons, and that this process requires nuclear-encoded protein translation, mediated by eEF1A1. Understanding how mitochondrial replication occurs in neurons, particularly at the level of the synapse, provides novel lines of research to tackle the pathological mechanisms underlying neurodegenerative diseases.
11h30 - 11h45 Oral communication (10-15 min)
Mitochondria damage-associated molecular patterns associated to extracellular microvesicles promote microglia-mediated proinflammatory response
Carla Lopes (CNC, CIBB & MIA, University of Coimbra, Portugal)
Click for Abstract
Mitochondria damage-associated molecular patterns associated to extracellular microvesicles promote microglia-mediated proinflammatory response
Henrique Tavares (1), Cláudia M. Deus (1), Margarida Beatriz (1), Paulo Oliveira(1), Carla Lopes (1,2)
1- CNC – Center for Neuroscience and Cell Biology, CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
2- Multidisciplinary Institute of Ageing, MIA – Portugal, University of Coimbra; Portugal
Mitochondrial and autophagy dysfunction play important roles in the pathogenesis of neurodegenerative diseases. Mitochondrial quality control mechanisms (QCM) are essential to maintain the mitochondrial structure and function. However, these mechanisms can be disrupted by the impairment of the mitochondrial-lysosomal axis [1]. Mitochondria-derived vesicles (MDVs) can dispose mildly damaged mitochondrial components in a complementary degradative pathway [2]. MDVs are particularly enriched in mitochondria-associated damage-associated molecular patterns (mitDAMPs) which can be released into the extracellular space as extracellular vesicles (EVs) and induce microglia-mediated immune response.
We here evaluated if an impairment of the mitochondrial-lysosomal axis, in normal human dermal fibroblasts or in fibroblasts from sporadic Parkinson Disease (PD), regulate the extracellular release of MDV and induce a proinflammatory microglial profile. Moreover, we evaluated if treating fibroblasts with AntiOxCIN4, a mitochondria-targeted antioxidant, can prevent this phenotype.
Microglia was stimulated with cell-free mitochondria, mtDNA, EVs and EV-isolated DNA from AntiOxCIN4-treated and non-treated fibroblasts.
Fibroblasts treated with FCCP/Chloroquine (CQ) showed increased mitochondrial fission and reactive oxygen species levels along with decreased mitochondrial biogenesis factors and mitochondrial DNA copies. Also, the inhibition of the mitochondrial-lysosomal axis resulted in an increase of EV secretion, with higher mtDNA copies and increased DNA oxidation (8-OHdG levels). An alteration in microglia transcriptional profile for several inflammatory factors (IL-1β, TNF-α, Arg1, and HMGB1) and NF-kβ activation was measured upon treatment with different stimuli. FCCP/CQ-treated fibroblasts displayed greater oxidative DNA damage and mediated the most significant mRNA transcriptional fold changes in microglia. AntiOxCIN4 reverted the mitochondrial abnormalities found in FCCP/CQ-treated cells and decreased the number of mtDNA copies associated to EVs. We observed an increase in the EVs-associated mtDNA copies in PD fibroblasts, although no alteration was observed in cells. Surprisingly, EVs isolated from PD fibroblasts treated with AntiOxCIN4 appear to induce a lower microglia proinflammatory response.
Our data highlighted how a dysfunctional mitochondrial-lysosomal axis may influence EVs role as a trigger for microglia activation, associating mitochondria dysfunction, EV secretion and mitochondria-driven immune responses.
European Regional Development Fund (ERDF), through the COMPETE2020–Operational Programme for Competitiveness and Internationalization and Portuguese national funds via FCT- Fundação para a Ciência e a Tecnologia, under project UIDB/04539/2020, UIDP/04539/2020 and LA/P/0058/2020.
1. Beatriz, M., et al., Journal of Extracellular Biology, 2022. 1(10): p. e65.
2. Soubannier, V., et al., Curr Biol, 2012. 22(2): p. 135-41.
11h45-12h15
Defective autophagy masquerading as mitochondrial disease
Michael Duchen (UCL Consortium for Mitochondrial Research, United Kingdom)
Click for Abstract
Defective autophagy masquerading as mitochondrial disease
Kritarth Singh and Michael R Duchen
UCL Consortium for Mitochondrial Research and Department of Cell and Developmental Biology, University College London, London UK
Vici Syndrome, a congenital disease of childhood is caused by mutations of a protein known as EPG5 [1], a protein which is important in mediating autophagosome/lysosome fusion – a key step in autophagy and mitophagy [2]. This multisystem disorder includes developmental defects involving the heart, CNS and immune system, but also involves progressive neurodegeneration and epilepsy. Most affected children do not survive into teenage years and there is no treatment. We have asked whether defective autophagy/mitophagy leads to significant mitochondrial dysfunction, to what extent this might contribute to the presentation and progression of the disease and whether the mechanisms involved might present novel therapeutic targets.
We have found that in fibroblasts from children with Vici syndrome, mitochondrial function is profoundly disordered, with impaired respiratory chain function and reduced membrane potential. We have also found that mitochondrial calcium handling is abnormal, with unexpected changes in the expression of components of the calcium uptake machinery that lead to mitochondrial calcium overload and increased vulnerability to opening of the permeability transition pore (mPTP). Remarkably, some children have been misdiagnosed with mitochondrial disease before genomic identification of an EPG5 mutation [3]. Indeed, Vici syndrome appears to a paradigm for an increasingly recognised group of neurodevelopmental and neurodegenerative disorders involving impaired autophagy [4]. It seems plausible that affected children may benefit from pharmacological interventions targeting mitochondrial function, especially the mPTP or autophagy, and preliminary data suggest that pharmacological stimulation of autophagy in the cell model leads to improved bioenergetic function.
References
1 - Byrne S, ……., Fanto M, Gautel M, Jungbluth H et al EPG5-related Vici syndrome: a paradigm of neurodevelopmental disorders with defective autophagy., Brain. 2016 Mar;139(Pt 3):765-81.
2 - Wang Z, Miao G, Xue X, Guo X, Yuan C, Wang Z, Zhang G, Chen Y, Feng D, Hu J, Zhang H. The Vici Syndrome Protein EPG5 Is a Rab7 Effector that Determines the Fusion Specificity of Autophagosomes with Late Endosomes/Lysosomes. Mol Cell. 2016 Sep 1;63(5):781-95.
3 - Deneubourg C, Ramm M, Smith LJ, Baron O, Singh K, Byrne SC, Duchen MR, Gautel M, Eskelinen EL, Fanto M, Jungbluth H The spectrum of neurodevelopmental, neuromuscular and neurodegenerative disorders due to defective autophagy. Autophagy. 2022 Mar;18(3):496-517.
4 - Balasubramaniam S, Riley LG, Vasudevan A, Cowley MJ, Gayevskiy V, Sue CM, Edwards C, Edkins E, Junckerstorff R, Kiraly-Borri C, Rowe P, Christodoulou J EPG5-Related Vici Syndrome: A Primary Defect of Autophagic Regulation with an Emerging Phenotype Overlapping with Mitochondrial Disorders. JIMD Rep. 2018;42:19-29. doi: 10.1007/8904_2017_71.
12h15
Poster Awards
+ Closing Session
Salão Nobre