Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nicol Birsa is active.

Publication


Featured researches published by Nicol Birsa.


Journal of Biological Chemistry | 2014

Lysine 27 Ubiquitination of the Mitochondrial Transport Protein Miro Is Dependent on Serine 65 of the Parkin Ubiquitin Ligase

Nicol Birsa; Rosalind Norkett; Tobias Wauer; Tycho E. T. Mevissen; Hsiu-Chuan Wu; Thomas Foltynie; Kailash P. Bhatia; Warren D. Hirst; David Komander; Helene Plun-Favreau; Josef T. Kittler

Background: Miro is a mitochondrial protein involved in mitochondrial trafficking. Results: Mitochondrial damage drives rapid Miro ubiquitination in a manner dependent on Ser-65 in Parkin; however, Miro degradation is delayed. Conclusion: Ubiquitination of Miro, rather than its degradation, could act as a signal for mitochondrial arrest and clearance. Significance: Disruption of the mitochondrial transport machinery could be implicated in Parkinson disease. Mitochondrial transport plays an important role in matching mitochondrial distribution to localized energy production and calcium buffering requirements. Here, we demonstrate that Miro1, an outer mitochondrial membrane (OMM) protein crucial for the regulation of mitochondrial trafficking and distribution, is a substrate of the PINK1/Parkin mitochondrial quality control system in human dopaminergic neuroblastoma cells. Moreover, Miro1 turnover on damaged mitochondria is altered in Parkinson disease (PD) patient-derived fibroblasts containing a pathogenic mutation in the PARK2 gene (encoding Parkin). By analyzing the kinetics of Miro1 ubiquitination, we further demonstrate that mitochondrial damage triggers rapid (within minutes) and persistent Lys-27-type ubiquitination of Miro1 on the OMM, dependent on PINK1 and Parkin. Proteasomal degradation of Miro1 is then seen on a slower time scale, within 2–3 h of the onset of ubiquitination. We find Miro ubiquitination in dopaminergic neuroblastoma cells is independent of Miro1 phosphorylation at Ser-156 but is dependent on the recently identified Ser-65 residue within Parkin that is phosphorylated by PINK1. Interestingly, we find that Miro1 can stabilize phospho-mutant versions of Parkin on the OMM, suggesting that Miro is also part of a Parkin receptor complex. Moreover, we demonstrate that Ser-65 in Parkin is critical for regulating Miro levels upon mitochondrial damage in rodent cortical neurons. Our results provide new insights into the ubiquitination-dependent regulation of the Miro-mediated mitochondrial transport machinery by PINK1/Parkin and also suggest that disruption of this regulation may be implicated in Parkinson disease pathogenesis.


Biochemical Society Transactions | 2013

Mitochondrial trafficking in neurons and the role of the Miro family of GTPase proteins

Nicol Birsa; Rosalind Norkett; Nathalie F. Higgs; Guillermo López-Doménech; Josef T. Kittler

Correct mitochondrial dynamics are essential to neuronal function. These dynamics include mitochondrial trafficking and quality-control systems that maintain a precisely distributed and healthy mitochondrial network, so that local energy demands or Ca2+-buffering requirements within the intricate architecture of the neuron can be met. Mitochondria make use of molecular machinery that couples these organelles to microtubule-based transport via kinesin and dynein motors, facilitating the required long-range movements. These motors in turn are associated with a variety of adaptor proteins allowing additional regulation of the complex dynamics demonstrated by these organelles. Over recent years, a number of new motor and adaptor proteins have been added to a growing list of components implicated in mitochondrial trafficking and distribution. Yet, there are major questions that remain to be addressed about the regulation of mitochondrial transport complexes. One of the core components of this machinery, the mitochondrial Rho GTPases Miro1 (mitochondrial Rho 1) and Miro2 have received special attention due to their Ca2+-sensing and GTPase abilities, marking Miro an exceptional candidate for co-ordinating mitochondrial dynamics and intracellular signalling pathways. In the present paper, we discuss the wealth of literature regarding Miro-mediated mitochondrial transport in neurons and recently highlighted involvement of Miro proteins in mitochondrial turnover, emerging as a key process affected in neurodegeneration.


Journal of Biological Chemistry | 2016

DISC1-dependent Regulation of Mitochondrial Dynamics Controls the Morphogenesis of Complex Neuronal Dendrites

Rosalind Norkett; Souvik Modi; Nicol Birsa; Talia A. Atkin; Davor Ivankovic; Manav Pathania; Svenja V. Trossbach; Carsten Korth; Warren D. Hirst; Josef T. Kittler

The DISC1 protein is implicated in major mental illnesses including schizophrenia, depression, bipolar disorder, and autism. Aberrant mitochondrial dynamics are also associated with major mental illness. DISC1 plays a role in mitochondrial transport in neuronal axons, but its effects in dendrites have yet to be studied. Further, the mechanisms of this regulation and its role in neuronal development and brain function are poorly understood. Here we have demonstrated that DISC1 couples to the mitochondrial transport and fusion machinery via interaction with the outer mitochondrial membrane GTPase proteins Miro1 and Miro2, the TRAK1 and TRAK2 mitochondrial trafficking adaptors, and the mitochondrial fusion proteins (mitofusins). Using live cell imaging, we show that disruption of the DISC1-Miro-TRAK complex inhibits mitochondrial transport in neurons. We also show that the fusion protein generated from the originally described DISC1 translocation (DISC1-Boymaw) localizes to the mitochondria, where it similarly disrupts mitochondrial dynamics. We also show by super resolution microscopy that DISC1 is localized to endoplasmic reticulum contact sites and that the DISC1-Boymaw fusion protein decreases the endoplasmic reticulum-mitochondria contact area. Moreover, disruption of mitochondrial dynamics by targeting the DISC1-Miro-TRAK complex or upon expression of the DISC1-Boymaw fusion protein impairs the correct development of neuronal dendrites. Thus, DISC1 acts as an important regulator of mitochondrial dynamics in both axons and dendrites to mediate the transport, fusion, and cross-talk of these organelles, and pathological DISC1 isoforms disrupt this critical function leading to abnormal neuronal development.


Neurobiology of Disease | 2016

Miro sculpts mitochondrial dynamics in neuronal health and disease.

Michael J. Devine; Nicol Birsa; Josef T. Kittler

Neurons are highly polarised cells with an elaborate and diverse cytoarchitecture. But this complex architecture presents a major problem: how to appropriately distribute metabolic resources where they are most needed within the cell. The solution comes in the form of mitochondria: highly dynamic organelles subject to a repertoire of trafficking, fission/fusion and quality control systems which work in concert to orchestrate a precisely distributed and healthy mitochondrial network. Mitochondria are critical for maintaining local energy supply and buffering Ca(2+) flux within neurons, and are increasingly recognised as being essential for healthy neuronal function. Mitochondrial movements are facilitated by their coupling to microtubule-based transport via kinesin and dynein motors. Adaptor proteins are required for this coupling and the mitochondrial Rho GTPases Miro1 and Miro2 are core components of this machinery. Both Miros have Ca(2+)-sensing and GTPase domains, and are therefore ideally suited to coordinating mitochondrial dynamics with intracellular signalling pathways and local energy turnover. In this review, we focus on Miros role in mediating mitochondrial transport in neurons, and the relevance of these mechanisms to neuronal health and disease.


Neurochemistry International | 2017

Ubiquitination at the mitochondria in neuronal health and disease

Christian Covill-Cooke; Jack H. Howden; Nicol Birsa; Josef T. Kittler

The preservation of mitochondrial function is of particular importance in neurons given the high energy requirements of action potential propagation and synaptic transmission. Indeed, disruptions in mitochondrial dynamics and quality control are linked to cellular pathology in neurodegenerative diseases, such as Alzheimers and Parkinsons disease. Here, we will discuss the role of ubiquitination by the E3 ligases: Parkin, MARCH5 and Mul1, and how they regulate mitochondrial homeostasis. Furthermore, given the role of Parkin and Mul1 in the formation of mitochondria-derived vesicles we give an overview of this area of mitochondrial homeostasis. We highlight how through the activity of these enzymes and MDV formation, multiple facets of mitochondrial biology can be regulated, ensuring the functionality of the mitochondrial network thus preserving neuronal health.


The EMBO Journal | 2018

Miro proteins coordinate microtubule‐ and actin‐dependent mitochondrial transport and distribution

Guillermo López-Doménech; Christian Covill-Cooke; Davor Ivankovic; Els F. Halff; David F. Sheehan; Rosalind Norkett; Nicol Birsa; Josef T. Kittler

In the current model of mitochondrial trafficking, Miro1 and Miro2 Rho‐GTPases regulate mitochondrial transport along microtubules by linking mitochondria to kinesin and dynein motors. By generating Miro1/2 double‐knockout mouse embryos and single‐ and double‐knockout embryonic fibroblasts, we demonstrate the essential and non‐redundant roles of Miro proteins for embryonic development and subcellular mitochondrial distribution. Unexpectedly, the TRAK1 and TRAK2 motor protein adaptors can still localise to the outer mitochondrial membrane to drive anterograde mitochondrial motility in Miro1/2 double‐knockout cells. In contrast, we show that TRAK2‐mediated retrograde mitochondrial transport is Miro1‐dependent. Interestingly, we find that Miro is critical for recruiting and stabilising the mitochondrial myosin Myo19 on the mitochondria for coupling mitochondria to the actin cytoskeleton. Moreover, Miro depletion during PINK1/Parkin‐dependent mitophagy can also drive a loss of mitochondrial Myo19 upon mitochondrial damage. Finally, aberrant positioning of mitochondria in Miro1/2 double‐knockout cells leads to disruption of correct mitochondrial segregation during mitosis. Thus, Miro proteins can fine‐tune actin‐ and tubulin‐dependent mitochondrial motility and positioning, to regulate key cellular functions such as cell proliferation.


bioRxiv | 2018

Miro ubiquitination is critical for efficient damage-induced PINK1/Parkin-mediated mitophagy

Guillermo López-Doménech; Christian Covill-Cooke; Jack H. Howden; Nicol Birsa; Corinne Morfill; Nicholas J. Brandon; Josef T. Kittler

Clearance of mitochondria following damage is critical for neuronal homeostasis. Here, we investigate the role of Miro proteins in mitochondrial turnover by the PINK1 / Parkin mitochondrial quality control system in vitro and in vivo. We find that upon mitochondrial damage, Miro is promiscuously ubiquitinated on multiple lysine residues. Combined knockout of both Miro1 and Miro2 or block of Miro ubiquitination and subsequent degradation, lead to slowed mitophagy. In cultured neurons, Miro1 knockout also leads to delayed Parkin translocation onto damaged mitochondria and reduced mitochondrial clearance. In vivo, postnatal knockout of Miro1 in hippocampus and cortex disrupts mitophagy and leads to a dramatic age dependent upregulation of the mitofusin mitochondrial fusion machinery. Fluorescence imaging of aged neurons conditionally knocked out for Miro1 and expressing mitoDendra to label mitochondria in vivo, reveals that Mfn1 / Mfn2 upregulation leads to enlarged and hyperfused somatic mitochondria. Our results provide new insights into the role of Miro in PINK1/Parkin dependent mitophagy and further suggest that disruption of this regulation may be implicated in human neurological pathology.


The EMBO Journal | 2018

Mice with endogenous TDP‐43 mutations exhibit gain of splicing function and characteristics of amyotrophic lateral sclerosis

Pietro Fratta; Prasanth Sivakumar; Jack Humphrey; Kitty Lo; Thomas Ricketts; Hugo Oliveira; Jose M Brito‐Armas; Bernadett Kalmar; Agnieszka Ule; Yichao Yu; Nicol Birsa; Cristian Bodo; Toby Collins; Alexander E. Conicella; Alan Mejia Maza; Alessandro Marrero‐Gagliardi; Michelle Stewart; Joffrey Mianné; Silvia Corrochano; Warren Emmett; Gemma F. Codner; Michael Groves; Ryutaro Fukumura; Yoichi Gondo; Mark F. Lythgoe; Erwin Pauws; Emma Peskett; Philip Stanier; Lydia Teboul; Martina Hallegger

TDP‐43 (encoded by the gene TARDBP) is an RNA binding protein central to the pathogenesis of amyotrophic lateral sclerosis (ALS). However, how TARDBP mutations trigger pathogenesis remains unknown. Here, we use novel mouse mutants carrying point mutations in endogenous Tardbp to dissect TDP‐43 function at physiological levels both in vitro and in vivo. Interestingly, we find that mutations within the C‐terminal domain of TDP‐43 lead to a gain of splicing function. Using two different strains, we are able to separate TDP‐43 loss‐ and gain‐of‐function effects. TDP‐43 gain‐of‐function effects in these mice reveal a novel category of splicing events controlled by TDP‐43, referred to as “skiptic” exons, in which skipping of constitutive exons causes changes in gene expression. In vivo, this gain‐of‐function mutation in endogenous Tardbp causes an adult‐onset neuromuscular phenotype accompanied by motor neuron loss and neurodegenerative changes. Furthermore, we have validated the splicing gain‐of‐function and skiptic exons in ALS patient‐derived cells. Our findings provide a novel pathogenic mechanism and highlight how TDP‐43 gain of function and loss of function affect RNA processing differently, suggesting they may act at different disease stages.


bioRxiv | 2017

The mitochondrial Rho-GTPase, Miro, is resident at peroxisomes and regulates peroxisomal trafficking and morphology

Christian Covill-Cooke; Guillermo López-Doménech; Nicol Birsa; Josef T. Kittler

Peroxisomes are essential for a number of cellular functions, including reactive oxygen species metabolism, fatty acid β-oxidation and lipid biosynthesis. To ensure optimal functionality of peroxisome-dependent processes throughout the cell they must be trafficked; however, peroxisomal transport remains poorly characterised. Here we show that Miro1 and Miro2, outer mitochondrial membrane proteins essential for mitochondrial trafficking, are also localised to peroxisomes. Peroxisomal localisation of Miro1 is negatively regulated by its first GTPase domain and is mediated by an interaction through its transmembrane domain with the peroxisomal-membrane protein chaperone, Pex19. By using Miro1/2 double knockout mouse embryonic fibroblasts (MEFs) we find that the loss of Miro1/2 leads to a significant reduction in short-range microtubule-independent peroxisomal motility. Additionally, Miro regulates peroxisomal size and morphology. Our results contribute to the fundamental understanding of peroxisomal trafficking and morphology, supporting a complex crosstalk between peroxisomal and mitochondrial biology.


Brain | 2018

TDP-43 mutations increase HNRNP A1-7B through gain of splicing function

Prasanth Sivakumar; Francesca De Giorgio; Agnieszka M Ule; Jacob Neeves; Remya R Nair; Matthew P Bentham; Nicol Birsa; Jack Humphrey; Vincent Plagnol; Abraham Acevedo-Arozena; Thomas J Cunningham; Elizabeth M. C. Fisher; Pietro Fratta

Collaboration


Dive into the Nicol Birsa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Komander

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Davor Ivankovic

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hsiu-Chuan Wu

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Jack H. Howden

University College London

View shared research outputs
Researchain Logo
Decentralizing Knowledge