Network


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

Hotspot


Dive into the research topics where Vanessa A. Morais is active.

Publication


Featured researches published by Vanessa A. Morais.


Embo Molecular Medicine | 2009

Parkinson's disease mutations in PINK1 result in decreased Complex I activity and deficient synaptic function

Vanessa A. Morais; Patrik Verstreken; Anne Roethig; Joél Smet; An Snellinx; Mieke Vanbrabant; Dominik Haddad; Christian Frezza; Wilhelm Mandemakers; Daniela Vogt-Weisenhorn; Rudy Van Coster; Wolfgang Wurst; Luca Scorrano; Bart De Strooper

Mutations of the mitochondrial PTEN (phosphatase and tensin homologue)‐induced kinase1 (PINK1) are important causes of recessive Parkinson disease (PD). Studies on loss of function and overexpression implicate PINK1 in apoptosis, abnormal mitochondrial morphology, impaired dopamine release and motor deficits. However, the fundamental mechanism underlying these various phenotypes remains to be clarified. Using fruit fly and mouse models we show that PINK1 deficiency or clinical mutations impact on the function of Complex I of the mitochondrial respiratory chain, resulting in mitochondrial depolarization and increased sensitivity to apoptotic stress in mammalian cells and tissues. In Drosophila neurons, PINK1 deficiency affects synaptic function, as the reserve pool of synaptic vesicles is not mobilized during rapid stimulation. The fundamental importance of PINK1 for energy maintenance under increased demand is further corroborated as this deficit can be rescued by adding ATP to the synapse. The clinical relevance of our observations is demonstrated by the fact that human wild type PINK1, but not PINK1 containing clinical mutations, can rescue Complex 1 deficiency. Our work suggests that Complex I deficiency underlies, at least partially, the pathogenesis of this hereditary form of PD. As Complex I dysfunction is also implicated in sporadic PD, a convergence of genetic and environmental causes of PD on a similar mitochondrial molecular mechanism appears to emerge.


Science | 2012

Vitamin K2 Is a Mitochondrial Electron Carrier That Rescues Pink1 Deficiency

Melissa Vos; Giovanni Esposito; Janaka N. Edirisinghe; Sven Vilain; Dominik Haddad; Jan R. Slabbaert; Stefanie Van Meensel; Onno Schaap; Bart De Strooper; R. Meganathan; Vanessa A. Morais; Patrik Verstreken

Keeping Mitochondria in the Pink Pink1 is a mitochondrial kinase, and loss of Pink1 function in flies and mice results in the accumulation of inefficient mitochondria. In a screen for modifiers of the Parkinson-associated gene, pink1, Vos et al. (p. 1306, published online 10 May; see the Perspective by Bhalerao and Clandinin) identified the fruit fly homolog of UBIAD1, “Heix.” UBIAD1 was localized in mitochondria and was able to convert vitamin K1 into vitamin K2/menaquinone (MK-n, n the number of prenylgroups). In bacteria, vitamin K2/MK-n acts as an electron carrier in the membrane and, similarly, in Drosophila, mitochondrial vitamin K2 appeared to act as an electron carrier to facilitate adenosine triphosphate production. Fruit flies that lack heix showed severe mitochondrial defects that could be rescued by administering vitamin K2. Adding vitamin K rescues fruit flies bearing a mutation in a Parkinson’s disease gene homolog. Human UBIAD1 localizes to mitochondria and converts vitamin K1 to vitamin K2. Vitamin K2 is best known as a cofactor in blood coagulation, but in bacteria it is a membrane-bound electron carrier. Whether vitamin K2 exerts a similar carrier function in eukaryotic cells is unknown. We identified Drosophila UBIAD1/Heix as a modifier of pink1, a gene mutated in Parkinson’s disease that affects mitochondrial function. We found that vitamin K2 was necessary and sufficient to transfer electrons in Drosophila mitochondria. Heix mutants showed severe mitochondrial defects that were rescued by vitamin K2, and, similar to ubiquinone, vitamin K2 transferred electrons in Drosophila mitochondria, resulting in more efficient adenosine triphosphate (ATP) production. Thus, mitochondrial dysfunction was rescued by vitamin K2 that serves as a mitochondrial electron carrier, helping to maintain normal ATP production.


Science | 2014

PINK1 Loss of Function Mutations Affect Mitochondrial Complex I Activity via NdufA10 Ubiquinone Uncoupling

Vanessa A. Morais; Dominik Haddad; Katleen Craessaerts; Pieter-Jan De Bock; Jef Swerts; Sven Vilain; Liesbeth Aerts; Lut Overbergh; Anne Grünewald; Philip Seibler; Christine Klein; Kris Gevaert; Patrik Verstreken; Bart De Strooper

In the PINK1 Pathogenic mutations in the kinase PINK1 are causally related to Parkinsons disease (PD). One hypothesis proposes that PINK1 regulates mitophagy—the clearance of dysfunctional mitochondria. A second hypothesis suggests that PINK1 has a direct effect on mitochondrial complex I, affecting the maintenance of the electron transport chain (ETC) resulting in decreased mitochondrial membrane potential and dysfunctional mitochondria. In support of the second hypothesis, Morais et al. (p. 203, published online 20 March) observed a complex I deficit in fibroblasts and neurons derived from induced pluripotent stem cells from PINK1 patients before any mitophagy was induced. The phosphoproteome of complex I in liver and brain from mice deficient for Pink1, compared to wild-type animals, revealed that Ser250 in complex I subunit NdufA10 was differentially phosphorylated. Ser250 is critically involved in the reduction of ubiquinone by complex I, explaining why Pink1 knockout mice, flies, and patient cell lines show decreased mitochondrial membrane potential. Synaptic defects in pink1 null mutant Drosophila could be rescued using phosphomimetic NdufA10. Mitochondria lacking a Parkinson’s disease–associated kinase harbor a functionally important phosphorylation defect. Under resting conditions, Pink1 knockout cells and cells derived from patients with PINK1 mutations display a loss of mitochondrial complex I reductive activity, causing a decrease in the mitochondrial membrane potential. Analyzing the phosphoproteome of complex I in liver and brain from Pink1−/− mice, we found specific loss of phosphorylation of serine-250 in complex I subunit NdufA10. Phosphorylation of serine-250 was needed for ubiquinone reduction by complex I. Phosphomimetic NdufA10 reversed Pink1 deficits in mouse knockout cells and rescued mitochondrial depolarization and synaptic transmission defects in pinkB9-null mutant Drosophila. Complex I deficits and adenosine triphosphate synthesis were also rescued in cells derived from PINK1 patients. Thus, this evolutionary conserved pathway may contribute to the pathogenic cascade that eventually leads to Parkinson’s disease in patients with PINK1 mutations.


Journal of Cell Science | 2007

A cell biological perspective on mitochondrial dysfunction in Parkinson disease and other neurodegenerative diseases.

Wim Mandemakers; Vanessa A. Morais; Bart De Strooper

Dysfunction of mitochondria is frequently proposed to be involved in neurodegenerative disease. Deficiencies in energy supply, free radical generation, Ca2+ buffering or control of apoptosis, could all theoretically contribute to progressive decline of the central nervous system. Parkinson disease illustrates how mutations in very different genes finally impinge directly or indirectly on mitochondrial function, causing subtle but finally fatal dysfunction of dopaminergic neurons. Neurons in general appear more sensitive than other cells to mutations in genes encoding mitochondrial proteins. Particularly interesting are mutations in genes such as Opa1, Mfn1 and Dnm1l, whose products are involved in the dynamic morphological alterations and subcellular trafficking of mitochondria. These indicate that mitochondrial dynamics are especially important for the long-term maintenance of the nervous system. The emerging evidence clearly demonstrates the crucial role of specific mitochondrial functions in maintaining neuronal circuit integrity.


Cancer Research | 2004

Role of the Human ST6GalNAc-I and ST6GalNAc-II in the Synthesis of the Cancer-Associated Sialyl-Tn Antigen

Nuno T. Marcos; Sandra Pinho; Catarina Grandela; Andrea Cruz; Bénédicte Samyn-Petit; Anne Harduin-Lepers; Raquel Almeida; Filipe Silva; Vanessa A. Morais; Júlia Costa; Jan Kihlberg; Henrik Clausen; Celso A. Reis

The Sialyl-Tn antigen (Neu5Acα2–6GalNAc-O-Ser/Thr) is highly expressed in several human carcinomas and is associated with carcinoma aggressiveness and poor prognosis. We characterized two human sialyltransferases, CMP-Neu5Ac:GalNAc-R α2,6-sialyltransferase (ST6GalNAc)-I and ST6GalNAc-II, that are candidate enzymes for Sialyl-Tn synthases. We expressed soluble recombinant hST6GalNAc-I and hST6GalNAc-II and characterized the substrate specificity of both enzymes toward a panel of glycopeptides, glycoproteins, and other synthetic glycoconjugates. The recombinant ST6GalNAc-I and ST6GalNAc-II showed similar substrate specificity toward glycoproteins and GalNAcα-O-Ser/Thr glycopeptides, such as glycopeptides derived from the MUC2 mucin and the HIVgp120. We also observed that the amino acid sequence of the acceptor glycopeptide contributes to the in vitro substrate specificity of both enzymes. We additionally established a gastric cell line, MKN45, stably transfected with the full length of either ST6GalNAc-I or ST6GalNAc-II and evaluated the carbohydrate antigens expression profile induced by each enzyme. MKN45 transfected with ST6GalNAc-I showed high expression of Sialyl-Tn, whereas MKN45 transfected with ST6GalNAc-II showed the biosynthesis of the Sialyl-6T structure [Galβ1–3 (Neu5Acα2–6)GalNAc-O-Ser/Thr]. In conclusion, although both enzymes show similar in vitro activities when Tn antigen alone is available, whenever both Tn and T antigens are present, ST6GalNAc-I acts preferentially on Tn antigen, whereas the ST6GalNAc-II acts preferentially on T antigen. Our results show that ST6GalNAc-I is the major Sialyl-Tn synthase and strongly support the hypothesis that the expression of the Sialyl-Tn antigen in cancer cells is due to ST6GalNAc-I activity.


Journal of Alzheimer's Disease | 2010

Mitochondria Dysfunction and Neurodegenerative Disorders: Cause or Consequence

Vanessa A. Morais; Bart De Strooper

Mitochondria are crucial regulators of energy metabolism and apoptotic pathways and have been closely linked to the pathogenesis of neurodegenerative disorders. In this review we mainly focus on mitochondrial dysfunction in two of the most prevalent neurological disorders: Alzheimers disease and Parkinsons disease. We discuss whether the role of mitochondria in those diseases should be considered primordial or secondary to other processes that eventually lead to neurodegeneration. In the case of Parkinsons disease, the role of mitochondria is quite clear and might be involved in the mechanism of this disorder. For Alzheimers disease, the evidence in favor of such a link is more indirect, and mitochondrial dysfunction likely occurs at a later stage of the disorder.


Journal of Biological Chemistry | 2003

The transmembrane domain region of nicastrin mediates direct interactions with APH-1 and the gamma-secretase complex.

Vanessa A. Morais; Adam S. Crystal; Donald S. Pijak; Dan Carlin; Júlia Costa; Virginia M.-Y. Lee; Robert W. Doms

Nicastrin (NCT) is a type I integral membrane protein that is one of the four essential components of the γ-secretase complex, a protein assembly that catalyzes the intramembranous cleavage of the amyloid precursor protein and Notch. Other γ-secretase components include presenilin-1 (PS1), APH-1, and PEN-2, all of which span the membrane multiple times. The mechanism by which NCT associates with the γ-secretase complex and regulates its activity is unclear. To avoid the misfolding phenotype often associated with introducing deletions or mutations into heavily glycosylated and disulfide-bonded proteins such as NCT, we produced chimeras between human (hNCT) and Caenorhabditis elegans NCT (ceNCT). Although ceNCT did not associate with human γ-secretase components, all of the ceNCT/hNCT chimeras interacted with γ-secretase components from human, C. elegans, or both, indicating that they folded correctly. A region at the C-terminal end of hNCT, encompassing the last 50 residues of its ectodomain, the transmembrane domain, and the cytoplasmic domain was important for mediating interactions with human PS1, APH-1, and PEN-2. This finding is consistent with the fact that the bulk of the γ-secretase complex proteins resides within the membrane, with relatively small extramembranous domains. Finally, hNCT associated with hAPH-1 in the absence of PS, consistent with NCT and APH-1 forming a subcomplex prior to association with PS1 and PEN-2 and indicating that the interactions between NCT with PS1 may be indirect or stabilized by the presence of APH-1.


PLOS Genetics | 2012

The yeast complex I equivalent NADH dehydrogenase rescues pink1 mutants.

Sven Vilain; Giovanni Esposito; Dominik Haddad; Onno Schaap; Mariya P. Dobreva; Melissa Vos; Stefanie Van Meensel; Vanessa A. Morais; Bart De Strooper; Patrik Verstreken

Pink1 is a mitochondrial kinase involved in Parkinsons disease, and loss of Pink1 function affects mitochondrial morphology via a pathway involving Parkin and components of the mitochondrial remodeling machinery. Pink1 loss also affects the enzymatic activity of isolated Complex I of the electron transport chain (ETC); however, the primary defect in pink1 mutants is unclear. We tested the hypothesis that ETC deficiency is upstream of other pink1-associated phenotypes. We expressed Saccaromyces cerevisiae Ndi1p, an enzyme that bypasses ETC Complex I, or sea squirt Ciona intestinalis AOX, an enzyme that bypasses ETC Complex III and IV, in pink1 mutant Drosophila and find that expression of Ndi1p, but not of AOX, rescues pink1-associated defects. Likewise, loss of function of subunits that encode for Complex I–associated proteins displays many of the pink1-associated phenotypes, and these defects are rescued by Ndi1p expression. Conversely, expression of Ndi1p fails to rescue any of the parkin mutant phenotypes. Additionally, unlike pink1 mutants, fly parkin mutants do not show reduced enzymatic activity of Complex I, indicating that Ndi1p acts downstream or parallel to Pink1, but upstream or independent of Parkin. Furthermore, while increasing mitochondrial fission or decreasing mitochondrial fusion rescues mitochondrial morphological defects in pink1 mutants, these manipulations fail to significantly rescue the reduced enzymatic activity of Complex I, indicating that functional defects observed at the level of Complex I enzymatic activity in pink1 mutant mitochondria do not arise from morphological defects. Our data indicate a central role for Complex I dysfunction in pink1-associated defects, and our genetic analyses with heterologous ETC enzymes suggest that Ndi1p-dependent NADH dehydrogenase activity largely acts downstream of, or in parallel to, Pink1 but upstream of Parkin and mitochondrial remodeling.


Biochimica et Biophysica Acta | 2008

Functional role of N-glycosylation from ADAM10 in processing, localization and activity of the enzyme.

Cristina Escrevente; Vanessa A. Morais; Sascha Keller; Cláudio M. Soares; Peter Altevogt; Júlia Costa

A disintegrin and metalloprotease 10 (ADAM10) is a type I transmembrane glycoprotein with four potential N-glycosylation sites (N267, N278, N439 and N551), that cleaves several plasma membrane proteins. In this work, ADAM10 was found to contain high-mannose and complex-type glycans. Individual N-glycosylation site mutants S269A, T280A, S441A, T553A were constructed, and results indicated that all sites were occupied. T280A was found to accumulate in the endoplasmic reticulum as the non-processed precursor of the enzyme. Furthermore, it exhibited only residual levels of metalloprotease activity in vivo towards the L1 cell adhesion molecule, as well as in vitro, using a ProTNF-alpha peptide as substrate. S441A showed increased ADAM10 susceptibility to proteolysis. Mutation of N267, N439 and N551 did not completely abolish enzyme activity, however, reduced levels were found. ADAM10 is sorted into secretory vesicles, the exosomes. Here, a fraction of ADAM10 from exosomes was found to contain more processed N-linked glycans than the cellular enzyme. In conclusion, N-glycosylation is crucial for ADAM10 processing and resistance to proteolysis, and results suggest that it is required for full-enzyme activity.


Molecular Cell | 2013

Mutations in the Intellectual Disability Gene Ube2a Cause Neuronal Dysfunction and Impair Parkin-Dependent Mitophagy

Dominik Haddad; Sven Vilain; Melissa Vos; Giovanni Esposito; Samer Matta; Vera M. Kalscheuer; Katleen Craessaerts; Maarten Leyssen; Rafaella M.P. Nascimento; Angela M. Vianna-Morgante; Bart De Strooper; Hilde Van Esch; Vanessa A. Morais; Patrik Verstreken

The prevalence of intellectual disability is around 3%; however, the etiology of the disease remains unclear in most cases. We identified a series of patients with X-linked intellectual disability presenting mutations in the Rad6a (Ube2a) gene, which encodes for an E2 ubiquitin-conjugating enzyme. Drosophila deficient for dRad6 display defective synaptic function as a consequence of mitochondrial failure. Similarly, mouse mRad6a (Ube2a) knockout and patient-derived hRad6a (Ube2a) mutant cells show defective mitochondria. Using in vitro and in vivo ubiquitination assays, we show that RAD6A acts as an E2 ubiquitin-conjugating enzyme that, in combination with an E3 ubiquitin ligase such as Parkin, ubiquitinates mitochondrial proteins to facilitate the clearance of dysfunctional mitochondria in cells. Hence, we identify RAD6A as a regulator of Parkin-dependent mitophagy and establish a critical role for RAD6A in maintaining neuronal function.

Collaboration


Dive into the Vanessa A. Morais's collaboration.

Top Co-Authors

Avatar

Bart De Strooper

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Júlia Costa

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Patrik Verstreken

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Dominik Haddad

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert W. Doms

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Melissa Vos

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Sven Vilain

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Donald S. Pijak

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge