Network


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

Hotspot


Dive into the research topics where Mariana E. G. de Araujo is active.

Publication


Featured researches published by Mariana E. G. de Araujo.


Nature | 2015

SLC38A9 is a component of the lysosomal amino acid sensing machinery that controls mTORC1

Manuele Rebsamen; Lorena Pochini; Taras Stasyk; Mariana E. G. de Araujo; Michele Galluccio; Richard K. Kandasamy; Berend Snijder; Astrid Fauster; Elena L. Rudashevskaya; Manuela Bruckner; Stefania Scorzoni; Przemyslaw A. Filipek; Kilian Huber; Johannes W. Bigenzahn; Leonhard X. Heinz; Claudine Kraft; Keiryn L. Bennett; Cesare Indiveri; Lukas A. Huber; Giulio Superti-Furga

Cell growth and proliferation are tightly linked to nutrient availability. The mechanistic target of rapamycin complex 1 (mTORC1) integrates the presence of growth factors, energy levels, glucose and amino acids to modulate metabolic status and cellular responses. mTORC1 is activated at the surface of lysosomes by the RAG GTPases and the Ragulator complex through a not fully understood mechanism monitoring amino acid availability in the lysosomal lumen and involving the vacuolar H+-ATPase. Here we describe the uncharacterized human member 9 of the solute carrier family 38 (SLC38A9) as a lysosomal membrane-resident protein competent in amino acid transport. Extensive functional proteomic analysis established SLC38A9 as an integral part of the Ragulator–RAG GTPases machinery. Gain of SLC38A9 function rendered cells resistant to amino acid withdrawal, whereas loss of SLC38A9 expression impaired amino-acid-induced mTORC1 activation. Thus SLC38A9 is a physical and functional component of the amino acid sensing machinery that controls the activation of mTOR.


Journal of Cell Biology | 2006

p14-MP1-MEK1 signaling regulates endosomal traffic and cellular proliferation during tissue homeostasis.

David Teis; Nicole Taub; Robert Kurzbauer; Diana Hilber; Mariana E. G. de Araujo; Miriam Erlacher; Martin Offterdinger; Andreas Villunger; Stephan Geley; Georg Bohn; Christoph Klein; Michael W. Hess; Lukas A. Huber

The extracellular signal-regulated kinase (ERK) cascade regulates proliferation, differentiation, and survival in multicellular organisms. Scaffold proteins regulate intracellular signaling by providing critical spatial and temporal specificity. The scaffold protein MEK1 (mitogen-activated protein kinase and ERK kinase 1) partner (MP1) is localized to late endosomes by the adaptor protein p14. Using conditional gene disruption of p14 in mice, we now demonstrate that the p14–MP1-MEK1 signaling complex regulates late endosomal traffic and cellular proliferation. This function its essential for early embryogenesis and during tissue homeostasis, as revealed by epidermis-specific deletion of p14. These findings show that endosomal p14–MP1-MEK1 signaling has a specific and essential function in vivo and, therefore, indicate that regulation of late endosomal traffic by extracellular signals is required to maintain tissue homeostasis.


Journal of Cell Biology | 2014

The late endosomal p14–MP1 (LAMTOR2/3) complex regulates focal adhesion dynamics during cell migration

Natalia Schiefermeier; Julia M. Scheffler; Mariana E. G. de Araujo; Taras Stasyk; Teodor Yordanov; Hannes L. Ebner; Martin Offterdinger; Sebastian Munck; Michael W. Hess; Sara A. Wickström; Anika Lange; Winfried Wunderlich; Reinhard Fässler; David Teis; Lukas A. Huber

Late endosomes locally regulate cell migration by transporting the p14–MP1 scaffold complex to the vicinity of focal adhesions.


Science | 2017

Crystal structure of the human lysosomal mTORC1 scaffold complex and its impact on signaling

Mariana E. G. de Araujo; Andreas Naschberger; Barbara G. Fürnrohr; Taras Stasyk; Theresia Dunzendorfer-Matt; Stefan Lechner; Stefan Welti; Leopold Kremser; Giridhar Shivalingaiah; Martin Offterdinger; Herbert Lindner; Lukas A. Huber; Klaus Scheffzek

Structure of human mTORC1 components The mTORC1 (mechanistic target of rapamycin complex 1) complex garners much attention as a signaling hub that coordinates input from growth-factor receptors and nutrient availability with metabolism and cell growth and proliferation. de Araujo et al. report the crystal structure of the LAMTOR (or “Ragulator”) complex that helps assemble mTORC1 at the lysosomal membrane for activation. The structure and functional studies reveal how LAMTOR1 wraps around the other subunits to hold them in place and interacts with the Rag guanosine triphosphatases in the complex. Science, this issue p. 377 Structural insights into nutrient and growth factor signaling at the lysosome. The LAMTOR [late endosomal and lysosomal adaptor and MAPK (mitogen-activated protein kinase) and mTOR (mechanistic target of rapamycin) activator] complex, also known as “Ragulator,” controls the activity of mTOR complex 1 (mTORC1) on the lysosome. The crystal structure of LAMTOR consists of two roadblock/LC7 domain–folded heterodimers wrapped and apparently held together by LAMTOR1, which assembles the complex on lysosomes. In addition, the Rag guanosine triphosphatases (GTPases) associated with the pentamer through their carboxyl-terminal domains, predefining the orientation for interaction with mTORC1. In vitro reconstitution and experiments with site-directed mutagenesis defined the physiological importance of LAMTOR1 in assembling the remaining components to ensure fidelity of mTORC1 signaling. Functional data validated the effect of two short LAMTOR1 amino acid regions in recruitment and stabilization of the Rag GTPases.


Traffic | 2015

Ultrastructural Morphometry Points to a New Role for LAMTOR2 in Regulating the Endo/Lysosomal System.

Georg F. Vogel; Hannes L. Ebner; Mariana E. G. de Araujo; Thomas Schmiedinger; Oliver Eiter; Haymo Pircher; Karin Gutleben; Barbara Witting; David Teis; Lukas A. Huber; Michael W. Hess

The late endosomal adaptor protein LAMTOR2/p14 is essential for tissue homeostasis by controlling MAPK and mTOR signaling, which in turn regulate cell growth and proliferation, migration and spreading. Moreover, LAMTOR2 critically controls architecture and function of the endocytic system, including epidermal growth factor receptor (EGFR) degradation in lysosomes, positioning of late endosomes and defense against intracellular pathogens. Here we describe the multifaceted ultrastructural phenotype of the endo/lysosomal system of LAMTOR2‐deficient mouse embryonic fibroblasts. Quantitative (immuno‐)electron microscopy of cryo‐fixed samples revealed significantly reduced numbers of recycling tubules emanating from maturing multivesicular bodies (MVB). Instead, a distinct halo of vesicles surrounded MVB, tentatively interpreted as detached, jammed recycling tubules. These morphological changes in LAMTOR2‐deficient cells correlated with the presence of growth factors (e.g. EGF), but were similarly induced in control cells by inactivating mTOR. Furthermore, proper transferrin receptor trafficking and recycling were apparently dependent on an intact LAMTOR complex. Finally, a severe imbalance in the relative proportions of endo/lysosomes was found in LAMTOR2‐deficient cells, resulting from increased amounts of mature MVB and (autophago)lysosomes. These observations suggest that the LAMTOR/Ragulator complex is required not only for maintaining the homeostasis of endo/lysosomal subpopulations but also contributes to the proper formation of MVB‐recycling tubules, and regulation of membrane/cargo recycling from MVB.


PLOS ONE | 2013

Polymorphisms in the Gene Regions of the Adaptor Complex LAMTOR2/LAMTOR3 and Their Association with Breast Cancer Risk

Mariana E. G. de Araujo; Gertraud Erhart; Katharina Buck; Elisabeth Müller-Holzner; Michael Hubalek; Heidelinde Fiegl; Daniele Campa; Federico Canzian; Ursula Eilber; Jenny Chang-Claude; Stefan Coassin; Margot Haun; Lyudmyla Kedenko; Bernhard Paulweber; Roland Reitsamer; Irmgard Himmel; Dieter Flesch-Janys; Claudia Lamina; Florian Kronenberg; Lukas A. Huber; Anita Kloss-Brandstätter

Background The late endosomal LAMTOR complex serves as a convergence point for both the RAF/MEK/ERK and the PI3K/AKT/mTOR pathways. Interestingly, both of these signalling cascades play a significant role in the aetiology of breast cancer. Our aim was to address the possible role of genetic polymorphisms in LAMTOR2 and LAMTOR3 as genetic risk factors for breast cancer. Methodology/Results We sequenced the exons and exon–intron boundaries of LAMTOR2 (p14) and LAMTOR3 (MP1) in 50 prospectively collected pairs of cancerous tissue and blood samples from breast cancer patients and compared their genetic variability. We found one single nucleotide polymorphism (SNP) in LAMTOR2 (rs7541) and two SNPs in LAMTOR3 (rs2298735 and rs148972953) in both tumour and blood samples, but no somatic mutations in cancerous tissues. In addition, we genotyped all three SNPs in 296 samples from the Risk Prediction of Breast Cancer Metastasis Study and found evidence of a genetic association between rs148972953 and oestrogen (ER) and progesterone receptor negative status (PR) (ER: OR = 3.60 (1.15–11.28); PR: OR = 4.27 (1.43–12.72)). However, when we additionally genotyped rs148972953 in the MARIE study including 2,715 breast cancer cases and 5,216 controls, we observed neither a difference in genotype frequencies between patients and controls nor was the SNP associated with ER or PR. Finally, all three SNPs were equally frequent in breast cancer samples and female participants (n = 640) of the population-based SAPHIR Study. Conclusions The identified polymorphisms in LAMTOR2 and LAMTOR3 do not seem to play a relevant role in breast cancer. Our work does not exclude a role of other not yet identified SNPs or that the here annotated polymorphism may in fact play a relevant role in other diseases. Our results underscore the importance of replication in association studies.


Journal of Biological Chemistry | 2013

Stability of the Endosomal Scaffold Protein LAMTOR3 Depends on Heterodimer Assembly and Proteasomal Degradation

Mariana E. G. de Araujo; Taras Stasyk; Nicole Taub; Hannes L. Ebner; Beatrix Fürst; Przemyslaw A. Filipek; Sabine Weys; Michael W. Hess; Herbert Lindner; Leopold Kremser; Lukas A. Huber

Background: LAMTOR3 is an endosomal MAPK scaffold recently shown also to modulate the mTORC1 pathway. Results: Association to LAMTOR2 and recruitment to the endosomal membrane stabilize LAMTOR3 and prevent the premature degradation of the protein by the proteasome. Conclusion: The ubiquitin-proteasome pathway and LAMTOR complex assembly tightly control LAMTOR3 abundance. Significance: Cells may modulate the dual ERK/mTORC1 activity of LAMTOR3 by tightly controlling its intracellular protein level. LAMTOR3 (MP1) and LAMTOR2 (p14) form a heterodimer as part of the larger Ragulator complex that is required for MAPK and mTOR1 signaling from late endosomes/lysosomes. Here, we show that loss of LAMTOR2 (p14) results in an unstable cytosolic monomeric pool of LAMTOR3 (MP1). Monomeric cytoplasmic LAMTOR3 is rapidly degraded in a proteasome-dependent but lysosome-independent manner. Mutational analyses indicated that the turnover of the protein is dependent on ubiquitination of several lysine residues. Similarly, other Ragulator subunits, LAMTOR1 (p18), LAMTOR4 (c7orf59), and LAMTOR5 (HBXIP), are degraded as well upon the loss of LAMTOR2. Thus the assembly of the Ragulator complex is monitored by cellular quality control systems, most likely to prevent aberrant signaling at the convergence of mTOR and MAPK caused by a defective Ragulator complex.


Journal of Cell Biology | 2017

LAMTOR/Ragulator is a negative regulator of Arl8b- and BORC-dependent late endosomal positioning

Przemyslaw A. Filipek; Mariana E. G. de Araujo; Georg F. Vogel; Cedric H. De Smet; Daniela Eberharter; Manuele Rebsamen; Elena L. Rudashevskaya; Leopold Kremser; Teodor Yordanov; Philipp Tschaikner; Barbara G. Fürnrohr; Stefan Lechner; Theresia Dunzendorfer-Matt; Klaus Scheffzek; Keiryn L. Bennett; Giulio Superti-Furga; Herbert Lindner; Taras Stasyk; Lukas A. Huber

Signaling from lysosomes controls cellular clearance and energy metabolism. Lysosomal malfunction has been implicated in several pathologies, including neurodegeneration, cancer, infection, immunodeficiency, and obesity. Interestingly, many functions are dependent on the organelle position. Lysosomal motility requires the integration of extracellular and intracellular signals that converge on a competition between motor proteins that ultimately control lysosomal movement on microtubules. Here, we identify a novel upstream control mechanism of Arl8b-dependent lysosomal movement toward the periphery of the cell. We show that the C-terminal domain of lyspersin, a subunit of BLOC-1–related complex (BORC), is essential and sufficient for BORC-dependent recruitment of Arl8b to lysosomes. In addition, we establish lyspersin as the linker between BORC and late endosomal/lysosomal adaptor and mitogen activated protein kinase and mechanistic target of rapamycin activator (LAMTOR) complexes and show that epidermal growth factor stimulation decreases LAMTOR/BORC association, thereby promoting BORC- and Arl8b-dependent lysosomal centrifugal transport.


European Heart Journal | 2017

A novel but frequent variant in LPA KIV-2 is associated with a pronounced Lp(a) and cardiovascular risk reduction

Stefan Coassin; Gertraud Erhart; Hansi Weissensteiner; Mariana E. G. de Araujo; Claudia Lamina; Sebastian Schönherr; Lukas Forer; Margot Haun; Jamie Lee Losso; Anna Köttgen; Konrad Schmidt; Gerd Utermann; Annette Peters; Christian Gieger; Konstantin Strauch; Armin Finkenstedt; Reto Bale; Heinz Zoller; Bernhard Paulweber; Kai-Uwe Eckardt; Alexander Hüttenhofer; Lukas A. Huber; Florian Kronenberg

Aims Lp(a) concentrations represent a major cardiovascular risk factor and are almost entirely controlled by one single locus (LPA). However, many genetic factors in LPA governing the enormous variance of Lp(a) levels are still unknown. Since up to 70% of the LPA coding sequence are located in a difficult to access hypervariable copy number variation named KIV-2, we hypothesized that it may contain novel functional variants with pronounced effects on Lp(a) concentrations. We performed a large scale mutation analysis in the KIV-2 using an extreme phenotype approach. Methods and Results We compiled an discovery set of 123 samples showing discordance between LPA isoform phenotype and Lp(a) concentrations and controls. Using ultra-deep sequencing, we identified a splice site variant (G4925A) in preferential association with the smaller LPA isoforms. Follow-up in a European general population (n = 2892) revealed an exceptionally high carrier frequency of 22.1% in the general population. The variant explains 20.6% of the Lp(a) variance in carriers of low molecular weight (LMW) apo(a) isoforms (P = 5.75e-38) and reduces Lp(a) concentrations by 31.3 mg/dL. Accordingly the odds ratio for cardiovascular disease was reduced from 1.39 [95% confidence interval (CI): 1.17–1.66, P = 1.89e-04] for wildtype LMW individuals to 1.19 [95%CI: 0.92; 1.56, P = 0.19] in LMW individuals who were additionally positive for G4925A. Functional studies point towards a reduction of splicing efficiency by this novel variant. Conclusion A highly frequent but until now undetected variant in the LPA KIV-2 region is strongly associated with reduced Lp(a) concentrations and reduced cardiovascular risk in LMW individuals.


PLOS ONE | 2014

LAMTOR2-Mediated Modulation of NGF/MAPK Activation Kinetics during Differentiation of PC12 Cells

Bettina Thauerer; Paul Voegele; Natascha Hermann-Kleiter; Nikolaus Thuille; Mariana E. G. de Araujo; Martin Offterdinger; Gottfried Baier; Lukas A. Huber; Gabriele Baier-Bitterlich

LAMTOR2 (p14), a part of the larger LAMTOR/Ragulator complex, plays a crucial role in EGF-dependent activation of p42/44 mitogen-activated protein kinases (MAPK, ERK1/2). In this study, we investigated the role of LAMTOR2 in nerve growth factor (NGF)-mediated neuronal differentiation. Stimulation of PC12 (rat adrenal pheochromocytoma) cells with NGF is known to activate the MAPK. Pharmacological inhibition of MEK1 as well as siRNA–mediated knockdown of both p42 and p44 MAPK resulted in inhibition of neurite outgrowth. Contrary to expectations, siRNA–mediated knockdown of LAMTOR2 effectively augmented neurite formation and neurite length of PC12 cells. Ectopic expression of a siRNA-resistant LAMTOR2 ortholog reversed this phenotype back to wildtype levels, ruling out nonspecific off-target effects of this LAMTOR2 siRNA approach. Mechanistically, LAMTOR2 siRNA treatment significantly enhanced NGF-dependent MAPK activity, and this effect again was reversed upon expression of the siRNA-resistant LAMTOR2 ortholog. Studies of intracellular trafficking of the NGF receptor TrkA revealed a rapid colocalization with early endosomes, which was modulated by LAMTOR2 siRNA. Inhibition of LAMTOR2 and concomitant destabilization of the remaining members of the LAMTOR complex apparently leads to a faster release of the TrkA/MAPK signaling module and nuclear increase of activated MAPK. These results suggest a modulatory role of the MEK1 adapter protein LAMTOR2 in NGF-mediated MAPK activation required for induction of neurite outgrowth in PC12 cells.

Collaboration


Dive into the Mariana E. G. de Araujo's collaboration.

Top Co-Authors

Avatar

Lukas A. Huber

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Taras Stasyk

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Michael W. Hess

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

David Teis

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Giorgia Lamberti

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Hannes L. Ebner

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Martin Offterdinger

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Georg F. Vogel

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Herbert Lindner

Innsbruck Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge