Network


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

Hotspot


Dive into the research topics where Luke M. Healy is active.

Publication


Featured researches published by Luke M. Healy.


Brain | 2015

Roles of microglia in brain development, tissue maintenance and repair

Mackenzie Michell-Robinson; Hanane Touil; Luke M. Healy; David R. Owen; Bryce A. Durafourt; Amit Bar-Or; Jack P. Antel; Craig S. Moore

The emerging roles of microglia are currently being investigated in the healthy and diseased brain with a growing interest in their diverse functions. In recent years, it has been demonstrated that microglia are not only immunocentric, but also neurobiological and can impact neural development and the maintenance of neuronal cell function in both healthy and pathological contexts. In the disease context, there is widespread consensus that microglia are dynamic cells with a potential to contribute to both central nervous system damage and repair. Indeed, a number of studies have found that microenvironmental conditions can selectively modify unique microglia phenotypes and functions. One novel mechanism that has garnered interest involves the regulation of microglial function by microRNAs, which has therapeutic implications such as enhancing microglia-mediated suppression of brain injury and promoting repair following inflammatory injury. Furthermore, recently published articles have identified molecular signatures of myeloid cells, suggesting that microglia are a distinct cell population compared to other cells of myeloid lineage that access the central nervous system under pathological conditions. Thus, new opportunities exist to help distinguish microglia in the brain and permit the study of their unique functions in health and disease.


JCI insight | 2016

Glioblastoma-infiltrated innate immune cells resemble M0 macrophage phenotype

Konrad Gabrusiewicz; Benjamin Rodriguez; Jun Wei; Yuuri Hashimoto; Luke M. Healy; Sourindra Maiti; Ginu Thomas; Shouhao Zhou; Qianghu Wang; Ahmed Elakkad; Brandon D. Liebelt; Nasser K. Yaghi; Ravesanker Ezhilarasan; Neal Huang; Jeffrey S. Weinberg; Sujit S. Prabhu; Ganesh Rao; Raymond Sawaya; Lauren A. Langford; Janet M. Bruner; Gregory N. Fuller; Amit Bar-Or; Wei Li; Rivka R. Colen; Michael A. Curran; Krishna P. Bhat; Jack P. Antel; Laurence J.N. Cooper; Erik P. Sulman; Amy B. Heimberger

Glioblastomas are highly infiltrated by diverse immune cells, including microglia, macrophages, and myeloid-derived suppressor cells (MDSCs). Understanding the mechanisms by which glioblastoma-associated myeloid cells (GAMs) undergo metamorphosis into tumor-supportive cells, characterizing the heterogeneity of immune cell phenotypes within glioblastoma subtypes, and discovering new targets can help the design of new efficient immunotherapies. In this study, we performed a comprehensive battery of immune phenotyping, whole-genome microarray analysis, and microRNA expression profiling of GAMs with matched blood monocytes, healthy donor monocytes, normal brain microglia, nonpolarized M0 macrophages, and polarized M1, M2a, M2c macrophages. Glioblastoma patients had an elevated number of monocytes relative to healthy donors. Among CD11b+ cells, microglia and MDSCs constituted a higher percentage of GAMs than did macrophages. GAM profiling using flow cytometry studies revealed a continuum between the M1- and M2-like phenotype. Contrary to current dogma, GAMs exhibited distinct immunological functions, with the former aligned close to nonpolarized M0 macrophages.


Frontiers in Immunology | 2016

Cytokine-Defined B Cell Responses as Therapeutic Targets in Multiple Sclerosis

Rui Li; Ayman Rezk; Luke M. Healy; Gillian Muirhead; Alexandre Prat; Jennifer L. Gommerman; Amit Bar-Or

Important antibody-independent pathogenic roles of B cells are emerging in autoimmune diseases, including multiple sclerosis (MS). The contrasting results of different treatments targeting B cells in patients (in spite of predictions of therapeutic benefits from animal models) call for a better understanding of the multiple roles that distinct human B cell responses likely play in MS. In recent years, both murine and human B cells have been identified with distinct functional properties related to their expression of particular cytokines. These have included regulatory (Breg) B cells (secreting interleukin (IL)-10 or IL-35) and pro-inflammatory B cells (secreting tumor necrosis factor α, LTα, IL-6, and granulocyte macrophage colony-stimulating factor). Better understanding of human cytokine-defined B cell responses is necessary in both health and diseases, such as MS. Investigation of their surface phenotype, distinct functions, and the mechanisms of regulation (both cell intrinsic and cell extrinsic) may help develop effective treatments that are more selective and safe. In this review, we focus on mechanisms by which cytokine-defined B cells contribute to the peripheral immune cascades that are thought to underlie MS relapses, and the impact of B cell-directed therapies on these mechanisms.


Journal of Cerebral Blood Flow and Metabolism | 2017

Pro-inflammatory activation of primary microglia and macrophages increases 18 kDa translocator protein expression in rodents but not humans:

David R. Owen; Nehal Narayan; Lisa Wells; Luke M. Healy; Erica Smyth; Eugenii A. Rabiner; Dylan A. Galloway; John B. Williams; Joshua Lehr; Harpreet Mandhair; Laura Peferoen; Peter C. Taylor; Sandra Amor; Jack P. Antel; Paul M. Matthews; Craig S. Moore

The 18kDa Translocator Protein (TSPO) is the most commonly used tissue-specific marker of inflammation in positron emission tomography (PET) studies. It is expressed in myeloid cells such as microglia and macrophages, and in rodent myeloid cells expression increases with cellular activation. We assessed the effect of myeloid cell activation on TSPO gene expression in both primary human and rodent microglia and macrophages in vitro, and also measured TSPO radioligand binding with 3H-PBR28 in primary human macrophages. As observed previously, we found that TSPO expression increases (∼9-fold) in rodent-derived macrophages and microglia upon pro-inflammatory stimulation. However, TSPO expression does not increase with classical pro-inflammatory activation in primary human microglia (fold change 0.85 [95% CI 0.58–1.12], p = 0.47). In contrast, pro-inflammatory activation of human monocyte-derived macrophages is associated with a reduction of both TSPO gene expression (fold change 0.60 [95% CI 0.45–0.74], p = 0.02) and TSPO binding site abundance (fold change 0.61 [95% CI 0.49–0.73], p < 0.0001). These findings have important implications for understanding the biology of TSPO in activated macrophages and microglia in humans. They are also clinically relevant for the interpretation of PET studies using TSPO targeting radioligands, as they suggest changes in TSPO expression may reflect microglial and macrophage density rather than activation phenotype.


Nature | 2018

Microglial control of astrocytes in response to microbial metabolites

Veit Rothhammer; Davis M. Borucki; Emily Tjon; Maisa C. Takenaka; Chun-Cheih Chao; Alberto Ardura-Fabregat; Kalil Alves de Lima; Cristina Gutiérrez-Vázquez; Patrick Hewson; Ori Staszewski; Manon Blain; Luke M. Healy; Tradite Neziraj; Matilde Borio; Michael A. Wheeler; Loic Lionel Dragin; David Laplaud; Jack P. Antel; Jorge Ivan Alvarez; Marco Prinz; Francisco J. Quintana

Microglia and astrocytes modulate inflammation and neurodegeneration in the central nervous system (CNS)1–3. Microglia modulate pro-inflammatory and neurotoxic activities in astrocytes, but the mechanisms involved are not completely understood4,5. Here we report that TGFα and VEGF-B produced by microglia regulate the pathogenic activities of astrocytes in the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis. Microglia-derived TGFα acts via the ErbB1 receptor in astrocytes to limit their pathogenic activities and EAE development. Conversely, microglial VEGF-B triggers FLT-1 signalling in astrocytes and worsens EAE. VEGF-B and TGFα also participate in the microglial control of human astrocytes. Furthermore, expression of TGFα and VEGF-B in CD14+ cells correlates with the multiple sclerosis lesion stage. Finally, metabolites of dietary tryptophan produced by the commensal flora control microglial activation and TGFα and VEGF-B production, modulating the transcriptional program of astrocytes and CNS inflammation through a mechanism mediated by the aryl hydrocarbon receptor. In summary, we identified positive and negative regulators that mediate the microglial control of astrocytes. Moreover, these findings define a pathway through which microbial metabolites limit pathogenic activities of microglia and astrocytes, and suppress CNS inflammation. This pathway may guide new therapies for multiple sclerosis and other neurological disorders.TGFα and VEGF-B produced by microglia regulate astrocyte function in the experimental autoimmune encephalomyelitis model of multiple sclerosis.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Sphingosine 1-phosphate receptor modulation suppresses pathogenic astrocyte activation and chronic progressive CNS inflammation

Veit Rothhammer; Jessica E. Kenison; Emily Tjon; Maisa C. Takenaka; Kalil Alves de Lima; Davis M. Borucki; Chun-Cheih Chao; Annabel Wilz; Manon Blain; Luke M. Healy; Jack P. Antel; Francisco J. Quintana

Significance Secondary progressive multiple sclerosis (SPMS) inflicts severe and irreversible disability on the affected individuals. Astrocytes are thought to play a central role in the pathogenesis of SPMS. Here, we demonstrate that Sphingosine-1-receptor (S1PR) modulation suppresses pathogenic astrocyte activation and disease progression in an animal model of SPMS. Using functional in vitro assays, we defined direct effects of S1PR modulation on murine and human astrocytes, as well as astrocyte-mediated effects on microglia and proinflammatory monocytes. Finally, in unbiased transcriptome-wide studies on human astrocytes, we identified candidate targets for the modulation of astrocyte function in SPMS. Collectively, this study sheds light on the pathogenesis of SPMS and evaluates the therapeutic value of S1PR modulation in an animal model of SPMS. Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease of the CNS that causes disability in young adults as a result of the irreversible accumulation of neurological deficits. Although there are potent disease-modifying agents for its initial relapsing-remitting phase, these therapies show limited efficacy in secondary progressive MS (SPMS). Thus, there is an unmet clinical need for the identification of disease mechanisms and potential therapeutic approaches for SPMS. Here, we show that the sphingosine 1-phosphate receptor (S1PR) modulator fingolimod (FTY720) ameliorated chronic progressive experimental autoimmune encephalomyelitis in nonobese diabetic mice, an experimental model that resembles several aspects of SPMS, including neurodegeneration and disease progression driven by the innate immune response in the CNS. Indeed, S1PR modulation by FTY720 in murine and human astrocytes suppressed neurodegeneration-promoting mechanisms mediated by astrocytes, microglia, and CNS-infiltrating proinflammatory monocytes. Genome-wide studies showed that FTY720 suppresses transcriptional programs associated with the promotion of disease progression by astrocytes. The study of the molecular mechanisms controlling these transcriptional modules may open new avenues for the development of therapeutic strategies for progressive MS.


Annals of clinical and translational neurology | 2016

Effects of fumarates on circulating and CNS myeloid cells in multiple sclerosis

Mackenzie Michell-Robinson; Craig S. Moore; Luke M. Healy; Lindsay A. Osso; Nika Zorko; Vladimir Grouza; Hanane Touil; Laurence Poliquin‐Lasnier; Anne‐Marie Trudelle; Paul S. Giacomini; Amit Bar-Or; Jack P. Antel

Dimethyl fumarate (DMF), a therapy for relapsing‐remitting multiple sclerosis (RRMS), is implicated as acting on inflammatory and antioxidant responses within both systemic immune and/or central nervous system (CNS) compartments. Orally administered DMF is rapidly metabolized to monomethyl fumarate (MMF). Our aim was to analyze the impact of fumarates on antiinflammatory and antioxidant profiles of human myeloid cells found in the systemic compartment (monocytes) and in the inflamed CNS (blood‐derived macrophages and brain‐derived microglia).


Journal of Immunology | 2016

MerTK Is a Functional Regulator of Myelin Phagocytosis by Human Myeloid Cells

Luke M. Healy; Gabrielle Perron; So-Yoon Won; Mackenzie Michell-Robinson; Ayman Rezk; Samuel K. Ludwin; Craig S. Moore; Jeffery A. Hall; Amit Bar-Or; Jack P. Antel

Multifocal inflammatory lesions featuring destruction of lipid-rich myelin are pathologic hallmarks of multiple sclerosis. Lesion activity is assessed by the extent and composition of myelin uptake by myeloid cells present in such lesions. In the inflamed CNS, myeloid cells are comprised of brain-resident microglia, an endogenous cell population, and monocyte-derived macrophages, which infiltrate from the systemic compartment. Using microglia isolated from the adult human brain, we demonstrate that myelin phagocytosis is dependent on the polarization state of the cells. Myelin ingestion is significantly enhanced in cells exposed to TGF-β compared with resting basal conditions and markedly reduced in classically activated polarized cells. Transcriptional analysis indicated that TGF-β–treated microglia closely resembled M0 cells. The tyrosine kinase phagocytic receptor MerTK was one of the most upregulated among a select number of differentially expressed genes in TGF-β–treated microglia. In contrast, MerTK and its known ligands, growth arrest-specific 6 and Protein S, were downregulated in classically activated cells. MerTK expression and myelin phagocytosis were higher in CNS-derived microglia than observed in monocyte-derived macrophages, both basally and under all tested polarization conditions. Specific MerTK inhibitors reduced myelin phagocytosis and the resultant anti-inflammatory biased cytokine responses for both cell types. Defining and modulating the mechanisms that regulate myelin phagocytosis has the potential to impact lesion and disease evolution in multiple sclerosis. Relevant effects would include enhancing myelin clearance, increasing anti-inflammatory molecule production by myeloid cells, and thereby permitting subsequent tissue repair.


Frontiers in Cellular Neuroscience | 2017

Divergent neuroinflammatory regulation of microglial TREM expression and involvement of NF-κB

Rosie Owens; Kathleen Grabert; Claire L. Davies; Alessio Alfieri; Jack P. Antel; Luke M. Healy; Barry W. McColl

The triggering receptor expressed on myeloid cells (TREM) family of proteins are cell surface receptors with important roles in regulation of myeloid cell inflammatory activity. In the central nervous system, TREM2 is implicated in further roles in microglial homeostasis, neuroinflammation and neurodegeneration. Different TREM receptors appear to have contrasting roles in controlling myeloid immune activity therefore the relative and co-ordinated regulation of their expression is important to understand but is currently poorly understood. We sought to determine how microglial TREM expression is affected under neuroinflammatory conditions in vitro and in vivo. Our data show that microglial Trem1 and Trem2 gene expression are regulated in an opposing manner by lipopolysaccharide (LPS) in vitro in both adult murine and human microglia. LPS caused a significant induction of Trem1 and a contrasting suppression of Trem2 expression. We also observed similar divergent Trem1 and Trem2 responses in vivo in response to acute brain inflammation and acute cerebral ischaemia. Our data show that inhibition of NF-κB activation prevents the LPS-induced alterations in both Trem1 and Trem2 expression in vitro indicating NF-κB as a common signaling intermediate controlling these divergent responses. Distinct patterns of microglial Trem1 induction and Trem2 suppression to different Toll-like receptor (TLR) ligands were also evident, notably with Trem1 induction restricted to those ligands activating TLRs signaling via TRIF. Our data show co-ordinated but divergent regulation of microglial TREM receptor expression with a central role for NF-κB. Neuroinflammatory conditions that alter the balance in TREM expression could therefore be an important influence on microglial inflammatory and homeostatic activity with implications for neuroinflammatory and neurodegenerative disease.


Nature Immunology | 2016

USP15 regulates type I interferon response and is required for pathogenesis of neuroinflammation

Sabrina Torre; Maria J Polyak; David Langlais; Nassima Fodil; J. Kennedy; Irena Radovanovic; Joanne Berghout; Gabriel André Leiva-Torres; Connie M Krawczyk; Subburaj Ilangumaran; Karen L. Mossman; Chen Liang; Klaus-Peter Knobeloch; Luke M. Healy; Jack P. Antel; Nathalie Arbour; Alexandre Prat; Jacek Majewski; Mark Lathrop; Silvia M. Vidal; Philippe Gros

Genes and pathways in which inactivation dampens tissue inflammation present new opportunities for understanding the pathogenesis of common human inflammatory diseases, including inflammatory bowel disease, rheumatoid arthritis and multiple sclerosis. We identified a mutation in the gene encoding the deubiquitination enzyme USP15 (Usp15L749R) that protected mice against both experimental cerebral malaria (ECM) induced by Plasmodium berghei and experimental autoimmune encephalomyelitis (EAE). Combining immunophenotyping and RNA sequencing in brain (ECM) and spinal cord (EAE) revealed that Usp15L749R-associated resistance to neuroinflammation was linked to dampened type I interferon responses in situ. In hematopoietic cells and in resident brain cells, USP15 was coexpressed with, and functionally acted together with the E3 ubiquitin ligase TRIM25 to positively regulate type I interferon responses and to promote pathogenesis during neuroinflammation. The USP15-TRIM25 dyad might be a potential target for intervention in acute or chronic states of neuroinflammation.

Collaboration


Dive into the Luke M. Healy's collaboration.

Top Co-Authors

Avatar

Jack P. Antel

Montreal Neurological Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar

Amit Bar-Or

Montreal Neurological Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar

Craig S. Moore

Memorial University of Newfoundland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

So-Yoon Won

Montreal Neurological Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar

Timothy E. Kennedy

Montreal Neurological Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar

Alexandre Prat

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar

Gabrielle Perron

Montreal Neurological Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar

Mackenzie Michell-Robinson

Montreal Neurological Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar

Manon Blain

Montreal Neurological Institute and Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge