Network


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

Hotspot


Dive into the research topics where Patrick Lajoie is active.

Publication


Featured researches published by Patrick Lajoie.


Journal of Cell Biology | 2009

Lattices, rafts, and scaffolds: domain regulation of receptor signaling at the plasma membrane

Patrick Lajoie; Jacky G. Goetz; James W. Dennis; Ivan R. Nabi

The plasma membrane is organized into various subdomains of clustered macromolecules. Such domains include adhesive structures (cellular synapses, substrate adhesions, and cell–cell junctions) and membrane invaginations (clathrin-coated pits and caveolae), as well as less well-defined domains such as lipid rafts and lectin-glycoprotein lattices. Domains are organized by specialized scaffold proteins including the intramembranous caveolins, which stabilize lipid raft domains, and the galectins, a family of animal lectins that cross-link glycoproteins forming molecular lattices. We review evidence that these heterogeneous microdomains interact to regulate substratum adhesion and cytokine receptor dynamics at the cell surface.


PLOS ONE | 2009

Screen for Chemical Modulators of Autophagy Reveals Novel Therapeutic Inhibitors of mTORC1 Signaling

Aruna D. Balgi; Bruno D. Fonseca; Elizabeth Donohue; Trevor C. F. Tsang; Patrick Lajoie; Christopher G. Proud; Ivan R. Nabi; Michel Roberge

Background Mammalian target of rapamycin complex 1 (mTORC1) is a protein kinase that relays nutrient availability signals to control numerous cellular functions including autophagy, a process of cellular self-eating activated by nutrient depletion. Addressing the therapeutic potential of modulating mTORC1 signaling and autophagy in human disease requires active chemicals with pharmacologically desirable properties. Methodology/Principal Findings Using an automated cell-based assay, we screened a collection of >3,500 chemicals and identified three approved drugs (perhexiline, niclosamide, amiodarone) and one pharmacological reagent (rottlerin) capable of rapidly increasing autophagosome content. Biochemical assays showed that the four compounds stimulate autophagy and inhibit mTORC1 signaling in cells maintained in nutrient-rich conditions. The compounds did not inhibit mTORC2, which also contains mTOR as a catalytic subunit, suggesting that they do not inhibit mTOR catalytic activity but rather inhibit signaling to mTORC1. mTORC1 inhibition and autophagosome accumulation induced by perhexiline, niclosamide or rottlerin were rapidly reversed upon drug withdrawal whereas amiodarone inhibited mTORC1 essentially irreversibly. TSC2, a negative regulator of mTORC1, was required for inhibition of mTORC1 signaling by rottlerin but not for mTORC1 inhibition by perhexiline, niclosamide and amiodarone. Transient exposure of immortalized mouse embryo fibroblasts to these drugs was not toxic in nutrient-rich conditions but led to rapid cell death by apoptosis in starvation conditions, by a mechanism determined in large part by the tuberous sclerosis complex protein TSC2, an upstream regulator of mTORC1. By contrast, transient exposure to the mTORC1 inhibitor rapamycin caused essentially irreversible mTORC1 inhibition, sustained inhibition of cell growth and no selective cell killing in starvation. Conclusion/Significance The observation that drugs already approved for human use can reversibly inhibit mTORC1 and stimulate autophagy should greatly facilitate the preclinical and clinical testing of mTORC1 inhibition for indications such as tuberous sclerosis, diabetes, cardiovascular disease and cancer.


Journal of Cellular and Molecular Medicine | 2007

Regulation of raft‐dependent endocytosis

Patrick Lajoie; Ivan R. Nabi

•  Introduction •  Raft‐dependent endocytosis encompasses various pathways •  Cav1 and the regulation of raft‐dependent endocytosis •  Signaling and raft‐dependent endocytosis •  Conclusion


Cancer and Metastasis Reviews | 2008

Caveolin-1 in tumor progression: the good, the bad and the ugly

Jacky G. Goetz; Patrick Lajoie; Sam M. Wiseman; Ivan R. Nabi

Caveolin-1 (Cav1) is a multifunctional scaffolding protein with multiple binding partners that is associated with cell surface caveolae and the regulation of lipid raft domains. Cav1 regulates multiple cancer-associated processes including cellular transformation, tumor growth, cell migration and metastasis, cell death and survival, multidrug resistance and angiogenesis. However, Cav1 has been reported to impact both positively and negatively on various aspects of tumor progression and while reported to function as a tumor suppressor, it has also been identified as a poor prognostic factor in various human cancers. In this review, we survey the functional roles of Cav1 in cancer and argue that Cav1 function is interdependent on tumor stage and the expression of molecular effectors that impact on its role during tumor progression.


International Review of Cell and Molecular Biology | 2010

Lipid rafts, caveolae, and their endocytosis.

Patrick Lajoie; Ivan R. Nabi

Lipid rafts are plasma membrane microdomains enriched in cholesterol and sphingolipids that are involved in the lateral compartmentalization of molecules at the cell surface. Internalization of ligands and receptors by these domains occurs via a process defined as raft-dependent endocytosis. Caveolae are caveolin-1-enriched smooth invaginations of the plasma membrane that form a subdomain of lipid rafts. Endocytosis of rafts, including caveolar but also noncaveolar dynamin-dependent and dynamin-independent pathways, is characterized by its cholesterol sensitivity and clathrin-independence. In this review we will characterize lipid rafts and caveolae, their endocytosis and its regulation by the actin cytoskeleton, caveolin-1, dynamin, and cholesterol.


Journal of Cell Biology | 2007

Plasma membrane domain organization regulates EGFR signaling in tumor cells

Patrick Lajoie; Emily A. Partridge; Ginette Guay; Jacky G. Goetz; Judy Pawling; Annick Lagana; Bharat H. Joshi; James W. Dennis; Ivan R. Nabi

Macromolecular complexes exhibit reduced diffusion in biological membranes; however, the physiological consequences of this characteristic of plasma membrane domain organization remain elusive. We report that competition between the galectin lattice and oligomerized caveolin-1 microdomains for epidermal growth factor (EGF) receptor (EGFR) recruitment regulates EGFR signaling in tumor cells. In mammary tumor cells deficient for Golgi β1,6N-acetylglucosaminyltransferase V (Mgat5), a reduction in EGFR binding to the galectin lattice allows an increased association with stable caveolin-1 cell surface microdomains that suppresses EGFR signaling. Depletion of caveolin-1 enhances EGFR diffusion, responsiveness to EGF, and relieves Mgat5 deficiency–imposed restrictions on tumor cell growth. In Mgat5+/+ tumor cells, EGFR association with the galectin lattice reduces first-order EGFR diffusion rates and promotes receptor interaction with the actin cytoskeleton. Importantly, EGFR association with the lattice opposes sequestration by caveolin-1, overriding its negative regulation of EGFR diffusion and signaling. Therefore, caveolin-1 is a conditional tumor suppressor whose loss is advantageous when β1,6GlcNAc-branched N-glycans are below a threshold for optimal galectin lattice formation.


Journal of Cell Biology | 2008

Concerted regulation of focal adhesion dynamics by galectin-3 and tyrosine-phosphorylated caveolin-1

Jacky G. Goetz; Bharat H. Joshi; Patrick Lajoie; Scott S. Strugnell; Trevor Scudamore; Liliana D. Kojic; Ivan R. Nabi

Both tyrosine-phosphorylated caveolin-1 (pY14Cav1) and GlcNAc-transferase V (Mgat5) are linked with focal adhesions (FAs); however, their function in this context is unknown. Here, we show that galectin-3 binding to Mgat5-modified N-glycans functions together with pY14Cav1 to stabilize focal adhesion kinase (FAK) within FAs, and thereby promotes FA disassembly and turnover. Expression of the Mgat5/galectin lattice alone induces FAs and cell spreading. However, FAK stabilization in FAs also requires expression of pY14Cav1. In cells lacking the Mgat5/galectin lattice, pY14Cav1 is not sufficient to promote FAK stabilization, FA disassembly, and turnover. In human MDA-435 cancer cells, Cav1 expression, but not mutant Y14FCav1, stabilizes FAK exchange and stimulates de novo FA formation in protrusive cellular regions. Thus, transmembrane crosstalk between the galectin lattice and pY14Cav1 promotes FA turnover by stabilizing FAK within FAs defining previously unknown, interdependent roles for galectin-3 and pY14Cav1 in tumor cell migration.


PLOS ONE | 2010

Formation and toxicity of soluble polyglutamine oligomers in living cells.

Patrick Lajoie; Erik L. Snapp

Background Aggregation and cytotoxicity of mutant proteins containing an expanded number of polyglutamine (polyQ) repeats is a hallmark of several diseases, including Huntingtons disease (HD). Within cells, mutant Huntingtin (mHtt) and other polyglutamine expansion mutant proteins exist as monomers, soluble oligomers, and insoluble inclusion bodies (IBs). Determining which of these forms constitute a toxic species has proven difficult. Recent studies support a role for IBs as a cellular coping mechanism to sequester levels of potentially toxic soluble monomeric and oligomeric species of mHtt. Methodology/Principal Findings When fused to a fluorescent reporter (GFP) and expressed in cells, the soluble monomeric and oligomeric polyglutamine species are visually indistinguishable. Here, we describe two complementary biophysical fluorescence microscopy techniques to directly detect soluble polyglutamine oligomers (using Htt exon 1 or Httex1) and monitor their fates in live cells. Photobleaching analyses revealed a significant reduction in the mobilities of mHttex1 variants consistent with their incorporation into soluble microcomplexes. Similarly, when fused to split-GFP constructs, both wildtype and mHttex1 formed oligomers, as evidenced by the formation of a fluorescent reporter. Only the mHttex1 split-GFP oligomers assembled into IBs. Both FRAP and split-GFP approaches confirmed the ability of mHttex1 to bind and incorporate wildtype Htt into soluble oligomers. We exploited the irreversible binding of split-GFP fragments to forcibly increase levels of soluble oligomeric mHttex1. A corresponding increase in the rate of IBs formation and the number formed was observed. Importantly, higher levels of soluble mHttex1 oligomers significantly correlated with increased mutant cytotoxicity, independent of the presence of IBs. Conclusions/Significance Our study describes powerful and sensitive tools for investigating soluble oligomeric forms of expanded polyglutamine proteins, and their impact on cell viability. Moreover, these methods should be applicable for the detection of soluble oligomers of a wide variety of aggregation prone proteins.


Molecular Biology of the Cell | 2010

Evolutionary Gain of Function for the ER Membrane Protein Sec62 from Yeast to Humans

Linda Müller; Maria Diaz de Escauriaza; Patrick Lajoie; Melanie Theis; Martin Jung; Anika Müller; Carsten Burgard; Markus Greiner; Erik L. Snapp; Johanna Dudek; Richard Zimmermann

We characterized interactions between the human proteins Sec62 and Sec63 as well as the putative interaction of human Sec62 with ribosomes. The data demonstrate evolutionary conservation of Sec62/Sec63 interaction and indicate that in the course of evolution Sec62 of vertebrates has gained the additional function to interact with ribosomes.


Journal of Cell Science | 2005

The lipid composition of autophagic vacuoles regulates expression of multilamellar bodies.

Patrick Lajoie; Ginette Guay; James W. Dennis; Ivan R. Nabi

Multilamellar bodies (MLBs) are responsible for surfactant secretion in type II alveolar cells but also accumulate in other cell types under pathological conditions, including cancer and lysosomal storage diseases such as Niemann-Pick C (NPC), a congenital disease where defective cholesterol transport leads to its accumulation in lysosomes. Mv1Lu type II alveolar cells transfected with Golgi β1,6 N-acetylglucosaminyltransferase V (Mgat5), enhancing the polylactosamine content of complex-type N-glycans, exhibit stable expression of MLBs whose formation requires lysosomal proteolysis within dense autophagic vacuoles. MLBs of Mgat5-transfected Mv1Lu cells are rich in phospholipids and have low levels of cholesterol. In Mv1Lu cells treated with the NPC-mimicking drug U18666A, cholesterol-rich MLBs accumulate independently of both Mgat5 expression and lysosomal proteolysis. Inhibition of autophagy by blocking the PI 3-kinase pathway with 3-methyladenine prevents MLB formation and results in the accumulation of non-lamellar, acidic lysosomal vacuoles. Treatment with 3-methyladenine inhibited the accumulation of monodansylcadaverine, a phospholipid-specific marker for autophagic vacuoles, but did not block endocytic access to the lysosomal vacuoles. Induction of autophagy via serum starvation resulted in an increased size of cholesterol-rich MLBs. Although expression of MLBs in the Mv1Lu cell line can be induced by modulating lysosomal cholesterol or protein glycosylation, an autophagic contribution of phospholipids is critical for the formation of concentric membrane lamellae within late lysosomal organelles.

Collaboration


Dive into the Patrick Lajoie's collaboration.

Top Co-Authors

Avatar

Erik L. Snapp

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ivan R. Nabi

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Jacky G. Goetz

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Bharat H. Joshi

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Liliana D. Kojic

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ginette Guay

Université de Montréal

View shared research outputs
Researchain Logo
Decentralizing Knowledge