Network


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

Hotspot


Dive into the research topics where Alexandre Patenaude is active.

Publication


Featured researches published by Alexandre Patenaude.


Microvascular Research | 2010

Involvement of endothelial progenitor cells in tumor vascularization

Alexandre Patenaude; Jeremy Parker; Aly Karsan

The generation of new microvasculature progresses by the process of angiogenesis, which involves the invasion and proliferation of endothelial cells from existing blood vessels into the local environment. In recent years, de novo generation of endothelial cells from circulating or local precursor endothelial cells is thought to also contribute to the neovasculature, a process that is referred to as vasculogenesis. In the adult, endothelial progenitor cells (EPC) are believed to be recruited from the bone marrow, migrate to sites requiring neovascularization and participate in the assembly of newly-forming blood vessels. A growing number of studies report that EPC participate in tumor progression and influence the efficacy of anticancer chemotherapeutics, and thus are attractive targets for cancer treatments. However, recent evidence calls into question the ability of marrow-derived EPC to act as a bona fide precursor for adult vasculogenesis. This review focuses on studies reporting or precluding the importance of EPC in tumor vasculogenesis. The putative sources of these cells and difficulties associated with their detection are discussed.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Differentiation of vascular smooth muscle cells from local precursors during embryonic and adult arteriogenesis requires Notch signaling

Linda Chang; Michela Noseda; Michelle Higginson; Michelle Ly; Alexandre Patenaude; Megan Fuller; Alastair H. Kyle; Andrew I. Minchinton; Mira C. Puri; Daniel J. Dumont; Aly Karsan

Vascular smooth muscle cells (VSMC) have been suggested to arise from various developmental sources during embryogenesis, depending on the vascular bed. However, evidence also points to a common subpopulation of vascular progenitor cells predisposed to VSMC fate in the embryo. In the present study, we use binary transgenic reporter mice to identify a Tie1+CD31dimvascular endothelial (VE)-cadherin−CD45− precursor that gives rise to VSMC in vivo in all vascular beds examined. This precursor does not represent a mature endothelial cell, because a VE-cadherin promoter-driven reporter shows no expression in VSMC during murine development. Blockade of Notch signaling in the Tie1+ precursor cell, but not the VE-cadherin+ endothelial cell, decreases VSMC investment of developing arteries, leading to localized hemorrhage in the embryo at the time of vascular maturation. However, Notch signaling is not required in the Tie1+ precursor after establishment of a stable artery. Thus, Notch activity is required in the differentiation of a Tie1+ local precursor to VSMC in a spatiotemporal fashion across all vascular beds.


Journal of Pharmacology and Experimental Therapeutics | 2006

Microtubule-Destabilizing Agents Induce Focal Adhesion Structure Disorganization and Anoikis in Cancer Cells

Réna G. Deschesnes; Alexandre Patenaude; Jean L.C. Rousseau; Jessica S. Fortin; Christine Ricard; Marie-France Côté; Jacques Huot; René C.-Gaudreault; Eric Petitclerc

Microtubule disruption provokes cytoskeleton and cell adhesion changes whose importance for apoptosis induction remains unclear. The present study focuses on the functional and the molecular adhesion kinetics that are induced by microtubule disruption-mediated apoptosis. We showed that antimicrotubules induce a biphasic sequence of adhesion response that precedes the onset of apoptosis and focal adhesion kinase hydrolysis. Antimicrotubules first induced an increase of the cellular adhesion paralleled by the raise of focal adhesion sites and actin contractility, which was followed by a sharp decrease of cell adhesion and disorganization of focal adhesion and actin stress fibers. The latter sequence of events ends by cell rounding, detachment from the extracellular matrix, and cell death. Microtubule-disrupting agents induced a sustained paxillin phosphorylation, before the activation of apoptosis, that requires the prior activation of extracellular signal-regulated kinase and p38 but not c-Jun NH2-terminal kinase. Interestingly, integrin-linked kinase overexpression rescued the antimicrotubule-mediated loss of cell viability. Altogether, these results propound that antimicrotubule agents induce anoikis through the loss of focal adhesion structure integrity.


Cancer Research | 2007

New Soft Alkylating Agents with Enhanced Cytotoxicity against Cancer Cells Resistant to Chemotherapeutics and Hypoxia

Alexandre Patenaude; Réna G. Deschesnes; Jean L.C. Rousseau; Eric Petitclerc; Jacques Lacroix; Marie-France Côté; René C.-Gaudreault

Chloroethylureas (CEU) are soft alkylating agents that covalently bind to beta-tubulin (betaTAC) and affect microtubule polymerization dynamics. Herein, we report the identification of a CEU subset and its corresponding oxazolines, which induce cell growth inhibition, apoptosis, and microtubule disruption without alkylating beta-tubulin (N-betaTAC). Both betaTAC and N-betaTAC trigger the collapse of mitochondrial potential (DeltaPsi(m)) and modulate reactive oxygen species levels, following activation of intrinsic caspase-8 and caspase-9. Experiments using human fibrosarcoma HT1080 respiratory-deficient cells (rho(0)) and uncoupler of the mitochondrial respiratory chain (MRC) showed that betaTAC and N-betaTAC impaired the MRC. rho(0) cells displayed an increased sensitivity toward N-betaTAC as compared with rho(+) cells but, in contrast, were resistant to betaTAC or classic chemotherapeutics, such as paclitaxel. Oxazoline-195 (OXA-195), an N-betaTAC derivative, triggered massive swelling of isolated mitochondria. This effect was insensitive to cyclosporin A and to Bcl-2 addition. In contrast, adenine nucleotide translocator (ANT) antagonists, bongkrekic acid or atractyloside, diminished swelling induced by OXA-195. The antiproliferative activities of the N-betaTACs CEU-025 and OXA-152 were markedly decreased in the presence of atractyloside. Conversely, pretreatment with cyclosporin A enhanced growth inhibition induced by betaTAC and N-betaTAC. One of the proteins alkylated by N-betaTAC was identified as the voltage-dependent anion channel isoform-1, an ANT partner. Our results suggest that betaTAC and N-betaTAC, despite their common ability to affect the microtubule network, trigger different cytotoxic mechanisms in cancer cells. The role of mitochondria in these mechanisms and the potential of N-betaTAC as a new therapeutic approach for targeting hypoxia-resistant cells are discussed.


Cancer Research | 2014

Endothelial-Specific Notch Blockade Inhibits Vascular Function and Tumor Growth through an eNOS-Dependent Mechanism

Alexandre Patenaude; Megan Fuller; Linda Chang; Fred Wong; Grigorios Paliouras; Rebecca Shaw; Alastair H. Kyle; Patricia Umlandt; Jennifer H.E. Baker; Erika Diaz; Jade Tong; Andrew I. Minchinton; Aly Karsan

Notch signaling is important for tumor angiogenesis induced by vascular endothelial growth factor A. Blockade of the Notch ligand Dll4 inhibits tumor growth in a paradoxical way. Dll4 inhibition increases endothelial cell sprouting, but vessels show reduced perfusion. The reason for this lack of perfusion is not currently understood. Here we report that inhibition of Notch signaling in endothelial cell using an inducible binary transgenic system limits VEGFA-driven tumor growth and causes endothelial dysfunction. Neither excessive endothelial cell sprouting nor defects of pericyte abundance accompanied the inhibition of tumor growth and functional vasculature. However, biochemical and functional analysis revealed that endothelial nitric oxide production is decreased by Notch inhibition. Treatment with the soluble guanylate cyclase activator BAY41-2272, a vasorelaxing agent that acts downstream of endothelial nitric oxide synthase (eNOS) by directly activating its soluble guanylyl cyclase receptor, rescued blood vessel function and tumor growth. We show that reduction in nitric oxide signaling is an early alteration induced by Notch inhibition and suggest that lack of functional vessels observed with Notch inhibition is secondary to inhibition of nitric oxide signaling. Coculture and tumor growth assays reveal that Notch-mediated nitric oxide production in endothelial cell requires VEGFA signaling. Together, our data support that eNOS inhibition is responsible for the tumor growth and vascular function defects induced by endothelial Notch inhibition. This study uncovers a novel mechanism of nitric oxide production in endothelial cells in tumors, with implications for understanding the peculiar character of tumor blood vessels.


Free Radical Biology and Medicine | 2002

Glutathione peroxidase-1 expression enhances recovery of human breast carcinoma cells from hyperoxic cell cycle arrest

Jean-François Bilodeau; Alexandre Patenaude; Bruno Piedboeuf; Caroline Carrier; Peter Petrov; Robert Faure; Marc-Edouard Mirault

We previously reported that hyperoxia (95% O(2)) induces an S-phase cell cycle arrest in glutathione peroxidase-deficient human carcinoma cells T47D-H3 (Exp. Cell Res. 256:347-357; 2000). Here, we investigated whether increasing the peroxide scavenging capacity via glutathione peroxidase-1 (GPx1) expression can prevent cell cycle alterations induced by oxidative stress. We show that GPx1-proficient T47D-GPx-2 transfectant cells, in which GPx1 concentration is most elevated in mitochondria (Biochem. Biophys. Res. Commun. 272:416-422; 2000), are partially resistant to cell cycle inhibition induced by hyperoxia or menadione exposure. Transient cell growth resistance was observed at the level of cell cycle phase distribution, Cdk2 activity, and DNA synthesis after 40 h hyperoxia. This differential resistance was associated with an inhibition of ROS production and lipid peroxidation induced by hyperoxia. After 64 h hyperoxic exposure, cell growth was completely abolished in both cell lines, despite elevated glutathione levels. However, in contrast to the GPx1-deficient cells, T47D-GPx-2 cells showed an increased capacity to recover from a cell cycle arrest mediated by a 64 h hyperoxic stress. Differential recovery was also observed at the ultrastructural level between Gpx1-proficient and -deficient cells. These data indicate that GPx1 played an important role in the cell capacity to recover from hyperoxic insults. The limited protection conferred by GPx1 during hyperoxia suggests that the deleterious effects were partially mediated by peroxide-derived free radicals, but also involved the action of nonperoxide-derived reactive species.


Bioorganic & Medicinal Chemistry Letters | 2008

Cycloalkyl-substituted aryl chloroethylureas inhibiting cell cycle progression in G0/G1 phase and thioredoxin-1 nuclear translocation

Jessica S. Fortin; Marie-France Côté; Jacques Lacroix; Alexandre Patenaude; Eric Petitclerc; René C.-Gaudreault

1-(2-Chloroethyl)-3-(4-cyclohexylphenyl)urea (cHCEU) has been shown to abrogate the presence of thioredoxin-1 into the nucleus through its selective covalent alkylation. In the present letter we have evaluated the structure-activity relationships of the substituents at positions 3 and 4 of the phenyl ring of cHCEU derivatives on cell cycle progression and thioredoxin-1 nuclear translocation. Active CEU derivatives exhibited GI(50) ranging from 1.9 to 49muM on breast carcinoma MCF-7, skin melanoma M21, and colon carcinoma HT-29 cells. On one hand, compounds 1, 2, 9c, 10c, 13, and 14 arrested the cell cycle in G(2)/M phase while CEUs 3, 4, 5c, 6c, 11c, and 12c blocked the cell division in G(0)/G(1) phase. On the other hand, CEUs 2-4, 5c, 7c, 8c, 11c, and 12c abrogated the translocation of thioredoxin-1 while the other CEU derivatives were inactive in that respect. Our results suggest that CEU substituted on the phenyl ring at position 3 or 4 by lower cycloalkyl or cycloalkoxy groups arrest cell progression in G(0)/G(1) phase through mechanism of action different from their antimicrotubule counterparts, presumably via thioredoxin-1 alkylation and modulation of its activity. The mechanism of action of these new molecules is still undetermined. However, the significant accumulation of cells in G(0)/G(1) phase suggests that these molecules may act similarly to known chemopreventive agents against cancers. In addition, the inhibition of Trx-1 nuclear localization also suggests the abrogation of an important chemoresistance mechanism towards a variety of chemotherapeutic agents.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2013

Notch-Dependent Regulation of the Ischemic Vasodilatory Response—Brief Report

Alex C.Y. Chang; Alexandre Patenaude; Katherine Lu; Megan Fuller; Michelle Ly; Alastair H. Kyle; Saeid Golbidi; Yingjin Wang; Keith Walley; Andrew I. Minchinton; Ismail Laher; Aly Karsan

Objective—We have recently described that Notch activates nitric oxide (NO) signaling in the embryonic endocardium. Both Notch signaling and NO signaling have been shown to be important during adult arteriogenesis. Notch has been shown to be required for remodeling of the collateral vessels, whereas NO is required for the initial vasodilatory response to ischemia. Whether Notch also has an impact on the vasodilatory phase of arteriogenesis after ischemia is not known. We tested the hypothesis that endothelial cell-Notch function is required for NO induction and vasodilation, in response to ischemia in the adult vasculature. Methods and Results—We observed a significant decrease in NO levels in the dorsal aorta using a mouse model where Notch was inhibited in endothelial cell in a Tet-inducible fashion. In a femoral artery ligation model, inhibition of endothelial cell-Notch reduced reperfusion and NO generation, as quantified by laser Doppler perfusion imaging and by phosphoendothelial NO synthase, nitrotyrosine, and phosphovasodilator-stimulated phosphoprotein staining, respectively. Conclusion—Endothelial Notch activation is required for NO production and reactive vasodilation in a femoral artery ligation model.


Microvascular Research | 2015

A novel population of local pericyte precursor cells in tumor stroma that require Notch signaling for differentiation

Alexandre Patenaude; Stefan Woerher; Patricia Umlandt; Fred Wong; Rawa Ibrahim; Alastair H. Kyle; Sandy Unger; Megan Fuller; Jeremy Parker; Andrew I. Minchinton; Connie J. Eaves; Aly Karsan

Pericytes are perivascular support cells, the origin of which in tumor tissue is not clear. Recently, we identified a Tie1(+) precursor cell that differentiates into vascular smooth muscle, in a Notch-dependent manner. To understand the involvement of Notch in the ontogeny of tumor pericytes we used a novel flow immunophenotyping strategy to define CD146(+)/CD45(-)/CD31(-/lo) pericytes in the tumor stroma. This strategy combined with ex vivo co-culture experiments identified a novel pericyte progenitor cell population defined as Sca1(hi)/CD146(-)/CD45(-)/CD31(-). The differentiation of these progenitor cells was stimulated by co-culture with endothelial cells. Overexpression of the Notch ligand Jagged1 in endothelial cells further stimulated the differentiation of Sca1(hi)/CD146(-)/CD45(-)/CD31(-) cells into pericytes, while inhibition of Notch signaling with a γ-secretase inhibitor reduced this differentiation. However, Notch inhibition specifically in Tie1-expressing cells did not change the abundance of pericytes in tumors, suggesting that the pericyte precursor is distinct from the vascular smooth muscle cell precursor. Transplant experiments showed that the bone marrow contributes minimally to tumor pericytes. Immunophenotyping revealed that Sca1(hi)/CD146(-)/CD45(-)/CD31(-) cells have greater potential to differentiate into pericytes and have increased expression of classic mesenchymal stem cell markers (CD13, CD44, Nt5e and Thy-1) compared to Sca1(-/lo)/CD146(-)/CD45(-)/CD31(-) cells. Our results suggest that a local Sca1(hi)/CD146(-)/CD45(-)/CD31(-) pericyte progenitor resides in the tumor microenvironment and requires Notch signaling for differentiation into mature pericytes.


Cancer Research | 2011

Abstract 3476: Blockade of endothelial Notch diminishes tumor growth and perfusion

Alexandre Patenaude; Linda Chang; Erika Diaz; Megan Fuller; Fred Wong; Aly Karsan

Canonical Notch signaling comprises a family of receptors and ligands that modulate cell fate and differentiation. Upon ligand binding, Notch receptors undergo a series of cleavages that culminate in the translocation of the intracellular domain to the nucleus, resulting in transcriptional activation through the derepression and activation at CSL-bound promoters. Notch activity is essential for embryonic vascular development and was recently also shown to be important for adult neovascularization. Notably, blocking Notch signaling by inhibition of the ligand Dll4 increased endothelial proliferation and sprouting, but resulted in non-perfused vasculature and tumor growth inhibition. Moreover, Dll4 inhibition was shown to diminish vascular mural cells, such as pericytes, in tumors. Whether these effects of Dll4 blockade on tumor vasculature resulted specifically from Notch inhibition in endothelial cells remains to be fully elucidated. We investigated this question using a tetracycline-inducible binary transgenic system, in which Notch is blocked by the expression of a dominant-negative form of mastermind-like 1 (dnMAML-GFP) under the control of the VE-cadherin promoter. Lewis lung carcinoma (LLC) cells overexpressing VEGFA (LLC-VEGFA) were implanted subcutaneously in the dorsa of double transgenic (Tg) or control mice. Tumors in the double Tg mice were significantly smaller than in control single Tg mice. Moreover, blood vessel perfusion from LLC-VEGFA tumors was significantly reduced in the double Tg mice compared to the control Tg mice. However, neither the abundance nor the branching architecture of LLC-VEGFA tumor blood vessels appeared to be different in the double Tg mice. Pericyte abundance, maturation and distribution relative to blood vessels did not change in the double Tg mice. Flow cytometry of tumor-derived cells and fluorescence microscopy analysis of tumor cryosections confirmed that dnMAML-GFP is expressed specifically in endothelial cells. Our data suggest that specific blockade of Notch in endothelial cells, induced by dnMAML-GFP, affects tumor growth through a mechanism different from previously-reported Dll4 inhibition studies. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3476. doi:10.1158/1538-7445.AM2011-3476

Collaboration


Dive into the Alexandre Patenaude's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aly Karsan

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Megan Fuller

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge