Network


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

Hotspot


Dive into the research topics where Daniel Canals is active.

Publication


Featured researches published by Daniel Canals.


Bioorganic & Medicinal Chemistry | 2009

Synthesis and biological properties of Pachastrissamine (jaspine B) and diastereoisomeric jaspines

Daniel Canals; David Mormeneo; Gemma Fabriàs; Amadeu Llebaria; Josefina Casas; Antonio Delgado

The synthesis of isomeric jaspines (anhydro phytosphingosines), arising from intramolecular cyclization of the corresponding phytosphingosines with different configurations at C3 and C4 positions of the sphingoid backbone, is reported. Natural jaspine B is the most cytotoxic isomer on A549 cells and it induces cell death in a dose-dependent manner. The cytotoxicity of jaspine B has been correlated with a significant increase of intracellular dihydroceramides, which seem to play an active role in autophagy.


FEBS Journal | 2013

Sphingosine-1-phosphate receptor 2.

Mohamad Adada; Daniel Canals; Yusuf A. Hannun; Lina M. Obeid

Sphingosine‐1‐phosphate (S1P) is a potent bioactive sphingolipid involved in cell proliferation, angiogenesis, inflammation and malignant transformation among other functions. S1P acts either directly on intracellular targets or activates G protein‐coupled receptors, specifically five S1P receptors (S1PRs). The identified S1PRs differ in cellular and tissue distribution, and each is coupled to specific G proteins, which mediate unique functions. Here, we describe functional characteristics of all five receptors, emphasizing S1PR2, which is critical in the immune, nervous, metabolic, cardiovascular, musculoskeletal, and renal systems. This review also describes the role of this receptor in tumor growth and metastasis and suggests potential therapeutic avenues that exploit S1PR2.


Analytical Chemistry | 2014

On-tissue localization of ceramides and other sphingolipids by MALDI mass spectrometry imaging.

E. Ellen Jones; Shaalee Dworski; Daniel Canals; Josefina Casas; Gemma Fabriàs; Drew Schoenling; Thierry Levade; Chadrick E. Denlinger; Yusuf A. Hannun; Jeffrey A. Medin; Richard R. Drake

A novel MALDI-FTICR imaging mass spectrometry (MALDI-IMS) workflow is described for on-tissue detection, spatial localization, and structural confirmation of low abundance bioactive ceramides and other sphingolipids. Increasingly, altered or elevated levels of sphingolipids, sphingolipid metabolites, and sphingolipid metabolizing enzymes have been associated with a variety of disorders such as diabetes, obesity, lysosomal storage disorders, and cancer. Ceramide, which serves as a metabolic hub in sphingolipid metabolism, has been linked to cancer signaling pathways and to metabolic regulation with involvement in autophagy, cell-cycle arrest, senescence, and apoptosis. Using kidney tissues from a new Farber disease mouse model in which ceramides of all acyl chain lengths and other sphingolipid metabolites accumulate in tissues, specific ceramides and sphingomyelins were identified by on-tissue isolation and fragmentation, coupled with an on-tissue digestion by ceramidase or sphingomyelinase. Multiple glycosphingolipid species were also detected. The newly generated library of sphingolipid ions was then applied to MALDI-IMS of human lung cancer tissues. Multiple tumor specific ceramide and sphingomyelin species were detected and confirmed by on-tissue enzyme digests and structural confirmation. High-resolution MALDI-IMS in combination with novel on-tissue ceramidase and sphingomyelinase enzyme digestions makes it now possible to rapidly visualize the distribution of bioactive ceramides and sphingomyelin in tissues.


Chemistry and Physics of Lipids | 2008

Cytotoxicity and acid ceramidase inhibitory activity of 2-substituted aminoethanol amides.

Carmen Bedia; Daniel Canals; Xavier Matabosch; Youssef Harrak; Josefina Casas; Amadeu Llebaria; Antonio Delgado; Gemma Fabriàs

The acid ceramidase (AC) inhibitory activity of octanoylamides, p-tert-butylbenzamides and pivaloylamides of several 2-substituted aminoethanols is reported. All the aminoethanol amides bearing a hexadecyl substituent (C16), as well as (S)-N-(1-(hexadecylthio)-3-hydroxypropan-2-yl)pivaloylamide (SC16-tb) were inhibitory in cell lysates overexpressing AC, while all other compounds were not inhibitors. Kinetic experiments with (R,E)-N-(1-hydroxyoctadec-3-en-2-yl)pivaloylamide (E-tb) and SC16-tb showed that inhibition was competitive, with K(i) values of 34 and 94.0 microM, respectively. None of the compounds inhibited neutral ceramidase. Compounds E-tb and E-c7 (the octanoylamide of the unsaturated base E), which elicited a dose-response inhibition with IC(50) values around 15 microM, were the only AC inhibitors in intact cells. Both compounds were toxic to A549 cells with LD(50) values nearly 40 microM. Flow cytometry studies with E-tb evidenced that this compound induced a concentration-dependent cell cycle arrest at G(1) and a 20-25% apoptosis/late apoptosis/necrosis after a 24-h incubation at 50 microM. In agreement with its activity as acidic ceramidase inhibitor, this effect was accompanied with an increase in the amounts of C14, C16 and C18 ceramides (LC-MS analyses), which suggested that these lipids may be responsible for the cytotoxic activity of E-tb.


ChemMedChem | 2007

Aminocyclitols as pharmacological chaperones for glucocerebrosidase, a defective enzyme in Gaucher disease.

Meritxell Egido-Gabás; Daniel Canals; Josefina Casas; Amadeu Llebaria; Antonio Delgado

3 pages, 4 figures.-- PMID: 17479993 [PubMed].-- Supporting information (11 pages) available at: http://www.wiley-vch.de/contents/jc_2452/2007/z700061_s.pdf


Biochemical Journal | 2013

Sphingosine 1-phosphate induces filopodia formation through S1PR2 activation of ERM proteins

K. Alexa Orr Gandy; Daniel Canals; Mohamad Adada; Masayuki Wada; Patrick Roddy; Ashley J. Snider; Yusuf A. Hannun; Lina M. Obeid

Previously we demonstrated that the sphingolipids ceramide and S1P (sphingosine 1-phosphate) regulate phosphorylation of the ERM (ezrin/radixin/moesin) family of cytoskeletal proteins [Canals, Jenkins, Roddy, Hernande-Corbacho, Obeid and Hannun (2010) J. Biol. Chem. 285, 32476-3285]. In the present article, we show that exogenously applied or endogenously generated S1P (in a sphingosine kinase-dependent manner) results in significant increases in phosphorylation of ERM proteins as well as filopodia formation. Using phosphomimetic and non-phosphorylatable ezrin mutants, we show that the S1P-induced cytoskeletal protrusions are dependent on ERM phosphorylation. Employing various pharmacological S1PR (S1P receptor) agonists and antagonists, along with siRNA (small interfering RNA) techniques and genetic knockout approaches, we identify the S1PR2 as the specific and necessary receptor to induce phosphorylation of ERM proteins and subsequent filopodia formation. Taken together, the results demonstrate a novel mechanism by which S1P regulates cellular architecture that requires S1PR2 and subsequent phosphorylation of ERM proteins.


Journal of Biological Chemistry | 2012

Protein phosphatase 1α mediates ceramide-induced ERM protein dephosphorylation: a novel mechanism independent of phosphatidylinositol 4, 5-biphosphate (PIP2) and myosin/ERM phosphatase.

Daniel Canals; Patrick Roddy; Yusuf A. Hannun

Background: ERM protein function is regulated by phosphorylation. Results: The PP1α isoform specifically dephosphorylates ceramide-induced ERM proteins. Conclusion: Plasma membrane ceramide activation of PP1α, a novel and a specific mechanism that regulates ERM proteins. Significance: The mechanisms of regulation of ERM proteins are important to understand and control cancer progression. ERM (ezrin, radixin, and moesin) proteins are cytoskeletal interacting proteins that bind cortical actin, the plasma membrane, and membrane proteins, which are found in specialized plasma membrane structures such as microvilli and filopodia. ERM proteins are regulated by phosphatidylinositol 4, 5-biphosphate (PIP2) and by phosphorylation of a C-terminal threonine, and its inactivation involves PIP2 hydrolysis and/or myosin phosphatase (MP). Recently, we demonstrated that ERM proteins are also subject to counter regulation by the bioactive sphingolipids ceramide and sphingosine 1-phosphate. Plasma membrane ceramide induces ERM dephosphorylation whereas sphingosine 1-phosphate induces their phosphorylation. In this work, we pursue the mechanisms by which ceramide regulates dephosphorylation. We found that this dephosphorylation was independent of hydrolysis and localization of PIP2 and MP. However, the results show that ERM dephosphorylation was blocked by treatment with protein phosphatase 1 (PP1) pharmacological inhibitors and specifically by siRNA to PP1α, whereas okadaic acid, a PP2A inhibitor, failed. Moreover, a catalytic inactive mutant of PP1α acted as dominant negative of the endogenous PP1α. Additional results showed that the ceramide mechanism of PP1α activation is largely independent of PIP2 hydrolysis and MP. Taken together, these results demonstrate a novel, acute mechanism of ERM regulation dependent on PP1α and plasma membrane ceramide.


Biochimica et Biophysica Acta | 2014

Sphingolipid regulation of ezrin, radixin, and moesin proteins family: implications for cell dynamics.

Mohamad Adada; Daniel Canals; Yusuf A. Hannun; Lina M. Obeid

A key but poorly studied domain of sphingolipid functions encompasses endocytosis, exocytosis, cellular trafficking, and cell movement. Recently, the ezrin, radixin and moesin (ERM) family of proteins emerged as novel potent targets regulated by sphingolipids. ERMs are structural proteins linking the actin cytoskeleton to the plasma membrane, also forming a scaffold for signaling pathways that are used for cell proliferation, migration and invasion, and cell division. Opposing functions of the bioactive sphingolipid ceramide and sphingosine-1-phosphate (S1P), contribute to ERM regulation. S1P robustly activates whereas ceramide potently deactivates ERM via phosphorylation/dephosphorylation, respectively. This recent dimension of cytoskeletal regulation by sphingolipids opens up new avenues to target cell dynamics, and provides further understanding of some of the unexplained biological effects mediated by sphingolipids. In addition, these studies are providing novel inroads into defining basic mechanisms of regulation and action of bioactive sphingolipids. This review describes the current understanding of sphingolipid regulation of the cytoskeleton, it also describes the biologies in which ERM proteins have been involved, and finally how these two large fields have started to converge. This article is part of a Special Issue entitled New Frontiers in Sphingolipid Biology.


Chemistry and Physics of Lipids | 2016

Inhibitors of the sphingomyelin cycle: Sphingomyelin synthases and sphingomyelinases.

Mohamad Adada; Chiara Luberto; Daniel Canals

Sphingolipids are a class of bioactive lipids, which are key modulators of an increasing number of physiologic and pathophysiologic processes that include cell cycle, apoptosis, angiogenesis, stress and inflammatory responses. Sphingomyelin is an important structural component of biological membranes, and one of the end-points in the synthesis of sphingolipids. Mainly synthetized in the Golgi apparatus, sphingomyelin is transported to all other biological membranes. Upon stimulation, sphingomyelin can be hydrolyzed to ceramide by 5 different sphingomyelinases. The diversity and cellular topology of ceramide allow it to exert multiple biologies. Furthermore, ceramide can be metabolized to many other bioactive sphingolipids. Ceramide, coming from sphingomyelin or other complex sphingolipids, can be hydrolyzed to sphingosine, which can easily change cellular localization. In turn, sphingosine can be recycled to ceramide and to sphingomyelin in the endoplasmic reticulum, completing the sphingomyelin cycle. Our understanding of the roles of various sphingolipids in the regulation of different cellular processes has come from studying the enzymes that regulate these sphingolipids, and their manipulation. The use of pharmacologic inhibitors has been critical for their study, as well as being promising bullets for disease treatment. Some of these diseases involving the sphingomyelin cycle include cancer, inflammation, atherosclerosis, diabetes and some rare diseases such as Niemann-Pick disease. This review will focus on the enzymes involved in the sphingomyelin cycle, their history, and their involvement in pathophysiological processes. Finally, it will describe in details all the small molecules that are being used to inhibit these enzymes and their use in therapeutics.


The FASEB Journal | 2015

Intracellular sphingosine kinase 2–derived sphingosine-1-phosphate mediates epidermal growth factor–induced ezrin-radixin-moesin phosphorylation and cancer cell invasion

Mohamad Adada; Daniel Canals; Nara Jeong; Ashwin D. Kelkar; María José Hernández-Corbacho; Michael J. Pulkoski-Gross; Jane C. Donaldson; Yusuf A. Hannun; Lina M. Obeid

The bioactive sphingolipid sphingosine‐1‐phosphate (S1P) mediates cellular proliferation, mitogenesis, inflammation, and angiogenesis. These biologies are mediated through S1P binding to specific GPCRs [sphingosine‐1‐phosphate receptor (S1PR)1‐5] and some other less well‐characterized intracellular targets. Ezrinradixin‐moesin (ERM) proteins, a family of adaptor molecules linking the cortical actin cytoskeleton to the plasma membrane, are emerging as critical regulators of cancer invasion via regulation of cell morphology and motility. Recently, we identified S1P as an acute ERM activator (via phosphorylation) through its action on S1PR2. In this work, we dissect the mechanism of S1P generation downstream of epidermal growth factor (EGF) leading to ERM phosphorylation and cancer invasion. Using pharmacologic inhibitors, small interfering RNA technologies, and genetic approaches, we demonstrate that sphingosine kinase (SK) 2, and not SK1, is essential and sufficient in EGF‐mediated ERM phosphorylation in HeLa cells. In fact, knocking down SK2 decreased ERM activation 2.5‐fold. Furthermore, we provide evidence that SK2 is necessary to mediate EGF‐induced invasion. In addition, overexpressing SK2 causes a 2‐fold increase in HeLa cell invasion. Surprisingly, and for the first time, we find that this event, although dependent on S1PR2 activation, does not generate and does not require extracellular S1P secretion, therefore introducing a potential novel model of autocrine/intracrine action of S1P that still involves its GPCRs. These results define new mechanistic insights for EGF‐mediated invasion and novel actions of SK2, therefore setting the stage for novel targets in the treatment of growth factor‐driven malignancies.—Adada, M. M., Canals, D., Jeong, N., Kelkar, A. D., Hernandez‐Corbacho, M., Pulkoski‐Gross, M. J., Donaldson, J. C., Hannun, Y. A., Obeid, L. M. Intracellular sphingosine kinase 2‐derived sphingosine‐1‐phosphate mediates epidermal growth factor‐induced ezrin‐radixin‐moesin phosphorylation and cancer cell invasion. FASEB J. 29, 4654‐4669 (2015). www.fasebj.org

Collaboration


Dive into the Daniel Canals's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Josefina Casas

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Amadeu Llebaria

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gemma Fabriàs

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

María José Hernández-Corbacho

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Patrick Roddy

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge