Network


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

Hotspot


Dive into the research topics where Maxime Hallé is active.

Publication


Featured researches published by Maxime Hallé.


Science Signaling | 2009

Leishmania GP63 Alters Host Signaling Through Cleavage-Activated Protein Tyrosine Phosphatases

Maria Adelaida Gomez; Irazú Contreras; Maxime Hallé; Michel L. Tremblay; R. McMaster; Martin Olivier

The parasite protein GP63 triggers cleavage and activation of host protein tyrosine phosphatases to promote infection. Exploiting the Host’s Phosphatases Leishmaniasis is a globally important infectious disease caused by the parasite Leishmania. Gomez et al. show that infection of macrophages with Leishmania alters the activity of multiple protein tyrosine phosphatases (PTPs) through cleavage mediated by the parasite protein GP63. The activated PTPs inhibit macrophage inflammatory immune responses through dephosphorylation of Janus kinases. In addition to the PTP SHP-1, previously reported to be activated in response to Leishmania infection, Gomez et al. show that the PTPs TCPTP and PTP1B are also activated and that PTP1B serves a key role in the initial stages of disease progression in mice. With more than 12 million people affected worldwide, 2 million new cases occurring per year, and the rapid emergence of drug resistance and treatment failure, leishmaniasis is an infectious disease for which research on drug and vaccine development, host-pathogen, and vector-parasite interactions are current international priorities. Upon Leishmania-macrophage interaction, activation of the protein tyrosine phosphatase (PTP) SHP-1 rapidly leads to the down-regulation of Janus kinase and mitogen-activated protein kinase signaling, resulting in the attenuation of host innate inflammatory responses and of various microbicidal macrophage functions. We report that, in addition to SHP-1, the PTPs PTP1B and TCPTP are activated and posttranslationally modified in infected macrophages, and we identify an essential role for PTP1B in the in vivo progression of Leishmania infection. The mechanism underlying PTP modulation involves the proteolytic activity of the Leishmania surface protease GP63. Access of GP63 to macrophage PTP1B, TCPTP, and SHP-1 is mediated in part by a lipid raft–dependent mechanism, resulting in PTP cleavage and stimulation of phosphatase activity. Collectively, our data present a mechanism of cleavage-dependent activation of macrophage PTPs by an obligate intracellular pathogen and show that internalization of GP63, a key Leishmania virulence factor, into host macrophages is a strategy the parasite uses to interact and survive within its host.


Molecular Cell | 2009

FAK Phosphorylation by ERK Primes Ras-Induced Tyrosine Dephosphorylation of FAK Mediated by PIN1 and PTP-PEST

Yanhua Zheng; Yan Xia; David H. Hawke; Maxime Hallé; Michel L. Tremblay; Xiang Gao; Xiao Zhen Zhou; Kenneth D. Aldape; Melanie H. Cobb; Keping Xie; Jie He; Zhimin Lu

Activated Ras has been found in many types of cancer. However, the mechanism underlying Ras-promoted tumor metastasis remains unclear. We demonstrate here that activated Ras induces tyrosine dephosphorylation and inhibition of FAK mediated by the Ras downstream Fgd1-Cdc42-PAK1-MEK-ERK signaling cascade. ERK phosphorylates FAK S910 and recruits PIN1 and PTP-PEST, which colocalize with FAK at the lamellipodia of migrating cells. PIN1 binding and prolyl isomerization of FAK cause PTP-PEST to interact with and dephosphorylate FAK Y397. Inhibition of FAK mediated by this signal relay promotes Ras-induced cell migration, invasion, and metastasis. These findings uncover the importance of sequential modification of FAK-by serine phosphorylation, isomerization, and tyrosine dephosphorylation--in the regulation of FAK activity and, thereby, in Ras-related tumor metastasis.


Journal of Biological Chemistry | 2009

The Leishmania Surface Protease GP63 Cleaves Multiple Intracellular Proteins and Actively Participates in p38 Mitogen-activated Protein Kinase Inactivation

Maxime Hallé; Maria Adelaida Gomez; Matthew Stuible; Hidehisa Shimizu; W. Robert McMaster; Martin Olivier; Michel L. Tremblay

The Leishmania parasite is a widespread disease threat in tropical areas, causing symptoms ranging from skin lesions to death. Leishmania parasites typically invade macrophages but are also capable of infecting fibroblasts, which may serve as a reservoir for recurrent infection. Invasion by intracellular pathogens often involves exploitation of the host cell cytoskeletal and signaling machinery. Here we have observed a dramatic rearrangement of the actin cytoskeleton and marked modifications in the profile of protein tyrosine phosphorylation in fibroblasts infected with Leishmania major. Correspondingly, exposure to L. major resulted in degradation of the phosphorylated adaptor protein p130Cas and the protein-tyrosine phosphatase-PEST. Cellular and in vitro assays using pharmacological protease inhibitors, recombinant enzyme, and genetically modified strains of L. major identified the parasite protease GP63 as the principal catalyst of proteolysis during infection. A number of additional signaling proteins were screened for degradation during L. major infection as follows: a small subset was cleaved, including cortactin, T-cell protein-tyrosine phosphatase, and caspase-3, but the majority remained unaffected. Protein degradation occurred in cells incubated with Leishmania extracts in the absence of intact parasites, suggesting a mechanism permitting transfer of functional GP63 into the intracellular space. Finally, we evaluated the impact of Leishmania on MAPK signaling; unlike p44/42 and JNK, p38 was inactivated upon infection in a GP63- and protein degradation-dependent manner, which likely involves cleavage of the upstream adaptor TAB1. Our results establish that GP63 plays a central role in a number of hostcell molecular events that likely contribute to the infectivity of Leishmania.


Molecular and Cellular Biology | 2007

Caspase-3 Regulates Catalytic Activity and Scaffolding Functions of the Protein Tyrosine Phosphatase PEST, a Novel Modulator of the Apoptotic Response

Maxime Hallé; Ying-Chih Liu; Serge Hardy; Jean-François Théberge; Christophe Blanchetot; Annie Bourdeau; Tzu-Ching Meng; Michel L. Tremblay

ABSTRACT The protein tyrosine phosphatase PEST (PTP-PEST) is involved in the regulation of the actin cytoskeleton. Despite the emerging functions attributed to both PTPs and the actin cytoskeleton in apoptosis, the involvement of PTP-PEST in apoptotic cell death remains to be established. Using several cell-based assays, we showed that PTP-PEST participates in the regulation of apoptosis. As apoptosis progressed, a pool of PTP-PEST localized to the edge of retracting lamellipodia. Expression of PTP-PEST also sensitized cells to receptor-mediated apoptosis. Concertedly, specific degradation of PTP-PEST was observed during apoptosis. Pharmacological inhibitors, immunodepletion experiments, and in vitro cleavage assays identified caspase-3 as the primary regulator of PTP-PEST processing during apoptosis. Caspase-3 specifically cleaved PTP-PEST at the 549DSPD motif and generated fragments, some of which displayed increased catalytic activity. Moreover, caspase-3 regulated PTP-PEST interactions with paxillin, leupaxin, Shc, and PSTPIP. PTP-PEST acted as a scaffolding molecule connecting PSTPIP to additional partners: paxillin, Shc, Csk, and activation of caspase-3 correlated with the modulation of the PTP-PEST adaptor function. In addition, cleavage of PTP-PEST facilitated cellular detachment during apoptosis. Together, our data demonstrate that PTP-PEST actively contributes to the cellular apoptotic response and reveal the importance of caspases as regulators of PTPs in apoptosis.


Cell Cycle | 2007

Protein tyrosine phosphatases: emerging regulators of apoptosis.

Maxime Hallé; Michel L. Tremblay; Tzu-Ching Meng

Apoptosis is a precisely controlled physiological mechanism that is required for the elimination of cells during embryonic development, in response to stress and infection as well as in the maintenance of homeostasis. Since the outcome of several of these biological processes is regulated by signaling events involving tyrosine phosphorylation, members of the protein tyrosine phosphatase (PTP) gene family are expected to be of primary importance. Here, we summarize the current literature linking the activities of classical PTPs with the regulation of apoptosis. The recent discovery of caspase-cleavage mediated modulation of a member of this family, PTP-PEST, indicates that other PTPs could be modulated in a similar manner. In light of this, we present an analysis of all murine and human PTPs gene for the presence of putative caspase cleavage motifs. Additional studies linking the activity of PTPs to their own regulation during programmed cell death initiation should provide important insight into the understanding of this fundamental physiological phenomenon.


Journal of Biological Chemistry | 2013

Regulation of the Src Kinase-associated Phosphoprotein 55 Homologue by the Protein Tyrosine Phosphatase PTP-PEST in the Control of Cell Motility

Emily Ayoub; Anita Hall; Adam M. Scott; Mélanie J. Chagnon; Géraldine Miquel; Maxime Hallé; Masaharu Noda; Andreas Bikfalvi; Michel L. Tremblay

Background: PTP-PEST regulates cell migration as part of many protein complexes. Results: SKAP-Hom is a substrate of PTP-PEST that is required for proper fibroblasts migration. Enhanced migration was observed when SKAP-Hom-deficient fibroblasts are rescued with its SH3 domain mutant. Conclusion: As a novel substrate of PTP-PEST, SKAP-Hom is important in cellular migration. Significance: PTP-PEST regulates migration through a new complex involving SKAP-Hom. PTP-PEST is a cytosolic ubiquitous protein tyrosine phosphatase (PTP) that contains, in addition to its catalytic domain, several protein-protein interaction domains that allow it to interface with several signaling pathways. Among others, PTP-PEST is a key regulator of cellular motility and cytoskeleton dynamics. The complexity of the PTP-PEST interactome underscores the necessity to identify its interacting partners and physiological substrates in order to further understand its role in focal adhesion complex turnover and actin organization. Using a modified yeast substrate trapping two-hybrid system, we identified a cytosolic adaptor protein named Src kinase-associated phosphoprotein 55 homologue (SKAP-Hom) as a novel substrate of PTP-PEST. To confirm PTP-PEST interaction with SKAP-Hom, in vitro pull down assays were performed demonstrating that the PTP catalytic domain and Proline-rich 1 (P1) domain are respectively binding to the SKAP-Hom Y260 and Y297 residues and its SH3 domain. Subsequently, we generated and rescued SKAP-Hom-deficient mouse embryonic fibroblasts (MEFs) with WT SKAP-Hom, SKAP-Hom tyrosine mutants (Y260F, Y260F/Y297F), or SKAP-Hom SH3 domain mutant (W335K). Given the role of PTP-PEST, wound-healing and trans-well migration assays were performed using the generated lines. Indeed, SKAP-Hom-deficient MEFs showed a defect in migration compared with WT-rescued MEFs. Interestingly, the SH3 domain mutant-rescued MEFs showed an enhanced cell migration corresponding potentially with higher tyrosine phosphorylation levels of SKAP-Hom. These findings suggest a novel role of SKAP-Hom and its phosphorylation in the regulation of cellular motility. Moreover, these results open new avenues by which PTP-PEST regulates cellular migration, a hallmark of metastasis.


Journal of Cell Science | 2016

PTP-PEST controls EphA3 activation and ephrin-induced cytoskeletal remodelling.

Mariam Mansour; Eva Nievergall; Kristina Gegenbauer; Carmen Llerena; Lakmali Atapattu; Maxime Hallé; Michel L. Tremblay; Peter W. Janes; Martin Lackmann

ABSTRACT Eph receptors and their corresponding membrane-bound ephrin ligands regulate cell positioning and establish tissue patterns during embryonic and oncogenic development. Emerging evidence suggests that assembly of polymeric Eph signalling clusters relies on cytoskeletal reorganisation and underlies regulation by protein tyrosine phosphatases (PTPs). PTP-PEST (also known as PTPN12) is a central regulator of actin cytoskeletal dynamics. Here, we demonstrate that an N-terminal fragment of PTP-PEST, generated through an ephrinA5-triggered and spatially confined cleavage mediated by caspase-3, attenuates EphA3 receptor activation and its internalisation. Isolation of EphA3 receptor signalling clusters within intact plasma membrane fragments obtained by detergent-free cell fractionation reveals that stimulation of cells with ephrin triggers effective recruitment of this catalytically active truncated form of PTP-PEST together with key cytoskeletal and focal adhesion proteins. Importantly, modulation of actin polymerisation using pharmacological and dominant-negative approaches affects EphA3 phosphorylation in a similar manner to overexpression of PTP-PEST. We conclude that PTP-PEST regulates EphA3 activation both by affecting cytoskeletal remodelling and through its direct action as a PTP controlling EphA3 phosphorylation, indicating its multifaceted regulation of Eph signalling. Highlighted Article: This study shows new roles for the protein tyrosine phosphatase PTP-PEST in controlling EphA3 receptor tyrosine kinase activity, trafficking and Eph-induced cell retraction.


Methods | 2005

Substrate-trapping techniques in the identification of cellular PTP targets.

Christophe Blanchetot; Mélanie J. Chagnon; N. Dubé; Maxime Hallé; Michel L. Tremblay


Journal of Biological Chemistry | 2002

PSTPIP is a substrate of PTP-PEST and serves as a scaffold guiding PTP-PEST toward a specific dephosphorylation of WASP.

Jean-François Côté; Ping Lin Chung; Jean-François Théberge; Maxime Hallé; Susan Spencer; Laurence A. Lasky; Michel L. Tremblay


Journal of Cell Science | 2005

Paxillin is essential for PTP-PEST-dependent regulation of cell spreading and motility: a role for paxillin kinase linker

Jennifer S. Jamieson; David A. Tumbarello; Maxime Hallé; Michael C. Brown; Michel L. Tremblay; Christopher E. Turner

Collaboration


Dive into the Maxime Hallé's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin Olivier

McGill University Health Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria Adelaida Gomez

McGill University Health Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

R. McMaster

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge