Network


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

Hotspot


Dive into the research topics where Huixian Gan is active.

Publication


Featured researches published by Huixian Gan.


Nature Immunology | 2009

Mycobacterium tuberculosis evades macrophage defenses by inhibiting plasma membrane repair

Maziar Divangahi; Minjian Chen; Huixian Gan; Danielle Desjardins; Tyler Hickman; David M. Lee; Sarah M. Fortune; Samuel M. Behar; Heinz G. Remold

Induction of macrophage necrosis is a strategy used by virulent Mycobacterium tuberculosis (Mtb) to avoid innate host defense. In contrast, attenuated Mtb causes apoptosis, which limits bacterial replication and promotes T cell cross-priming by antigen-presenting cells. Here we show that Mtb infection causes plasma membrane microdisruptions. Resealing of these lesions, a process crucial for preventing necrosis and promoting apoptosis, required translocation of lysosomal and Golgi apparatus–derived vesicles to the plasma membrane. Plasma membrane repair depended on prostaglandin E2 (PGE2), which regulates synaptotagmin 7 (Syt-7), the calcium sensor involved in the lysosome-mediated repair mechanism. By inducing production of lipoxin A4 (LXA4), which blocks PGE2 biosynthesis, virulent Mtb prevented membrane repair and induced necrosis. Thus, virulent Mtb impairs macrophage plasma membrane repair to evade host defenses.


Journal of Experimental Medicine | 2008

Lipid mediators in innate immunity against tuberculosis: opposing roles of PGE2 and LXA4 in the induction of macrophage death

Minjian Chen; Maziar Divangahi; Huixian Gan; Daniel S.J. Shin; Song Hong; David M. Lee; Charles N. Serhan; Samuel M. Behar; Heinz G. Remold

Virulent Mycobacterium tuberculosis (Mtb) induces a maladaptive cytolytic death modality, necrosis, which is advantageous for the pathogen. We report that necrosis of macrophages infected with the virulent Mtb strains H37Rv and Erdmann depends on predominant LXA4 production that is part of the antiinflammatory and inflammation-resolving action induced by Mtb. Infection of macrophages with the avirulent H37Ra triggers production of high levels of the prostanoid PGE2, which promotes protection against mitochondrial inner membrane perturbation and necrosis. In contrast to H37Ra infection, PGE2 production is significantly reduced in H37Rv-infected macrophages. PGE2 acts by engaging the PGE2 receptor EP2, which induces cyclic AMP production and protein kinase A activation. To verify a role for PGE2 in control of bacterial growth, we show that infection of prostaglandin E synthase (PGES)−/− macrophages in vitro with H37Rv resulted in significantly higher bacterial burden compared with wild-type macrophages. More importantly, PGES−/− mice harbor significantly higher Mtb lung burden 5 wk after low-dose aerosol infection with virulent Mtb. These in vitro and in vivo data indicate that PGE2 plays a critical role in inhibition of Mtb replication.


Mucosal Immunology | 2011

Apoptosis is an innate defense function of macrophages against Mycobacterium tuberculosis

Samuel M. Behar; Constance J. Martin; Matthew G. Booty; T. Nishimura; Xiaomin Zhao; Huixian Gan; Maziar Divangahi; Heinz G. Remold

Two different forms of death are commonly observed when Mycobacterium tuberculosis (Mtb)-infected macrophages die: (i) necrosis, a death modality defined by cell lysis and (ii) apoptosis, a form of death that maintains an intact plasma membrane. Necrosis is a mechanism used by bacteria to exit the macrophage, evade host defenses, and spread. In contrast, apoptosis of infected macrophages is associated with diminished pathogen viability. Apoptosis occurs when tumor necrosis factor activates the extrinsic death domain pathway, leading to caspase-8 activation. In addition, mitochondrial outer membrane permeabilization leading to activation of the intrinsic apoptotic pathway is required. Both pathways lead to caspase-3 activation, which results in apoptosis. We have recently demonstrated that during mycobacterial infection, cell death is regulated by the eicosanoids, prostaglandin E2 (proapoptotic) and lipoxin (LX)A4 (pronecrotic). Although PGE2 protects against necrosis, virulent Mtb induces LXA4 and inhibits PGE2 production. Under such conditions, mitochondrial inner membrane damage leads to macrophage necrosis. Thus, virulent Mtb subverts eicosanoid regulation of cell death to foil innate defense mechanisms of the macrophage.


Journal of Immunology | 2006

A Mechanism of Virulence: Virulent Mycobacterium tuberculosis Strain H37Rv, but Not Attenuated H37Ra, Causes Significant Mitochondrial Inner Membrane Disruption in Macrophages Leading to Necrosis

Minjian Chen; Huixian Gan; Heinz G. Remold

Infection of human monocyte-derived macrophages with Mycobacterium tuberculosis at low multiplicities of infection leads 48–72 h after the infection to cell death with the characteristics of apoptosis or necrosis. Predominant induction of one or the other cell death modality depends on differences in mitochondrial membrane perturbation induced by attenuated and virulent strains. Infection of macrophages with the attenuated H37Ra or the virulent H37Rv causes mitochondrial outer membrane permeabilization characterized by cytochrome c release from the mitochondrial intermembrane space and apoptosis. Mitochondrial outer membrane permeabilization is transient, peaks 6 h after infection, and requires Ca2+ flux and B cell chronic lymphocytic leukemia/lymphoma 2-associated protein X translocation into mitochondria. In contrast, only the virulent H37Rv induces significant mitochondrial transmembrane potential (Δψm) loss caused by mitochondrial permeability transition. Dissipation of Δψm also peaks at 6 h after infection, is transient, is inhibited by the classical mitochondrial permeability transition inhibitor cyclosporine A, has a requirement for mitochondrial Ca2+ loading, and is independent of B cell chronic lymphocytic leukemia/lymphoma translocation into the mitochondria. Transient dissipation of Δψm 6 h after infection is essential for the induction of macrophage necrosis by Mtb, a mechanism that allows further dissemination of the pathogen and development of the disease.


Nature Immunology | 2008

Mycobacterium tuberculosis blocks crosslinking of annexin-1 and apoptotic envelope formation on infected macrophages to maintain virulence

Huixian Gan; Jinhee Lee; Fucheng Ren; Minjian Chen; Hardy Kornfeld; Heinz G. Remold

Macrophages infected with attenuated Mycobacterium tuberculosis strain H37Ra become apoptotic, which limits bacterial replication and facilitates antigen presentation. Here we demonstrate that cells infected with H37Ra became apoptotic after the formation of an apoptotic envelope on their surface was complete. This process required exposure of phosphatidylserine on the cell surface, followed by deposition of the phospholipid-binding protein annexin-1 and then transglutaminase-mediated crosslinking of annexin-1 through its amino-terminal domain. In macrophages infected with the virulent strain H37Rv, in contrast, the amino-terminal domain of annexin-1 was removed by proteolysis, thus preventing completion of the apoptotic envelope, which resulted in macrophage death by necrosis. Virulent M. tuberculosis therefore avoids the host defense system by blocking formation of the apoptotic envelope, which leads to macrophage necrosis and dissemination of infection in the lung.


Journal of Immunology | 2001

Cytosolic Phospholipase A2 Participates with TNF-α in the Induction of Apoptosis of Human Macrophages Infected with Mycobacterium tuberculosis H37Ra

Lei Duan; Huixian Gan; Jonathan P. Arm; Heinz G. Remold

Macrophage (MΦ) apoptosis, an important innate microbial defense mechanism induced by Mycobacterium tuberculosis (Mtb) H37Ra, depends on the induction of TNF-α synthesis. When protein synthesis is blocked, both infection with Mtb and addition of TNF-α are required to induce caspase 9 activation, caspase 3 activation and apoptosis. In this study, we show that the second protein synthesis-independent signal involves activation of group IV cytosolic phospholipase A2 (cPLA2). Apoptosis of Mtb-infected MΦ and concomitant arachidonic acid release are abrogated by group IV cPLA2 inhibitors (methyl arachidonyl fluorophosphate and methyl trifluoromethyl ketone), but not by inhibitors of group VI Ca2+-independent (iPLA2 ; bromoenol lactone) or of secretory low molecular mass PLA2. In MΦ homogenates, the predominant PLA2 activity showed the same inhibitor sensitivity pattern and preferred arachidonic acid over palmitic acid in substrates, also indicating the presence of one or more group IV cPLA2 enzymes. In concordance with these findings, MΦ lysates contained transcripts and protein for group IV cPLA2-α and cPLA2-γ. Importantly, group IV cPLA2 inhibitors significantly reduced MΦ antimycobacterial activity and addition of arachidonic acid, the major product of group IV cPLA2, to infected MΦ treated with cPLA2 inhibitors completely restored the antimycobacterial activity. Importantly, addition of arachidonic acid alone to infected MΦ significantly reduced the mycobacterial burden. These findings indicate that Mtb induces MΦ apoptosis by independent signaling through at least two pathways, TNF-α and cPLA2, which are both also critical for antimycobacterial defense of the MΦ .


Journal of Immunology | 2002

Critical Role of Mitochondrial Damage in Determining Outcome of Macrophage Infection with Mycobacterium tuberculosis

Lei Duan; Huixian Gan; David E. Golan; Heinz G. Remold

Human macrophages (Mφ) respond to Mycobacterium tuberculosis (Mtb) infection by undergoing apoptosis, a cornerstone of effective antimycobacterial host defense. Virulent mycobacteria override this reaction by inducing necrosis leading to uncontrolled Mtb replication. Accordingly, Mφ death induced by inoculation with Mtb had the characteristics of apoptosis and necrosis and correlated with moderate increase of mitochondrial permeability transition (MPT), mitochondrial cytochrome c release, and caspase-9 and -3 activation. We hypothesized that changes in intramitochondrial Ca2+ concentration ([Ca2+]m) determine whether Mφ undergo either apoptosis or necrosis. Therefore, we induced mechanism(s) leading to predominant apoptosis or necrosis by modulating [Ca2+]m and examined their physiological consequences. Adding calcium ionophore A23187 to Mφ inoculated with Mtb further increased calcium flux into the cells which is thought to lead to increased [Ca2+]m, blocked necrosis, stabilized MPT, decreased mitochondrial cytochrome c release, lowered caspase activation, and accompanied effective antimycobacterial activity. In contrast, Mφ infected with Mtb in presence of the mitochondrial calcium uniporter inhibitor ruthenium red showed increased mitochondrial swelling and cytochrome c release and decreased MPT and antimycobacterial activity. Thus, in Mtb-infected Mφ, high levels of mitochondrial membrane integrity, low levels of caspase activation, and diminished mitochondrial cytochrome c release are hallmarks of apoptosis and effective antimycobacterial activity. In contrast, breakdown of mitochondrial membrane integrity and increased caspase activation are characteristic of necrosis and uncontrolled Mtb replication.


The Journal of Infectious Diseases | 2005

Enhancement of Antimycobacterial Activity of Macrophages by Stabilization of Inner Mitochondrial Membrane Potential

Huixian Gan; Xianbao He; Lei Duan; Elizabeth Mirabile-Levens; Hardy Kornfeld; Heinz G. Remold

Infection of human macrophages with Mycobacterium tuberculosis leads to cell death that, depending on the M. tuberculosis strain, time course, and multiplicity of infection, may have predominant features of apoptosis or necrosis. A key feature of infection-induced necrosis is mitochondrial damage characterized by an irreversible increase in the mitochondrial permeability transition (MPT), which is associated with increased release of cytochrome c from the mitochondria and uncontrolled mycobacterial replication. In contrast, protection of the mitochondria from MPT favors apoptosis of M. tuberculosis-infected macrophages. Apoptosis of M. tuberculosis-infected macrophages is associated with killing of intracellular M. tuberculosis, and this may be enhanced when MPT is stabilized. Here, we show that cyclosporin A (CsA), an inhibitor of MPT, protects the mitochondria from release of cytochrome c and promotes the antimycobacterial activity of macrophages infected with M. tuberculosis H37Ra. Signaling by purinergic P2 receptors has previously been linked to the antimycobacterial activity of macrophages. In the present study, we found that infection with H37Ra inhibits P2X7 receptor (P2XR) signals and that CsA restores P2XR function in infected macrophages. Together, these data demonstrate that CsA promotes at least 2 antimycobacterial pathways of macrophages.


The FASEB Journal | 2013

The prostaglandin E2 receptor EP4 is integral to a positive feedback loop for prostaglandin E2 production in human macrophages infected with Mycobacterium tuberculosis

Tomoyasu Nishimura; Xiaomin Zhao; Huixian Gan; Shigeo Koyasu; Heinz G. Remold

Prostaglandin E2 (PGE2) is an important biological mediator involved in the defense against Mycobacterium tuberculosis (Mtb) infection. Previously, we reported that in macrophages (Mφs), infection with avirulent Mtb H37Ra resulted in inhibition of necrosis by an inhibitory effect on mitochondrial permeability transition via the PGE2 receptor EP2. However, human Mφs also express EP4, a PGE2 receptor functionally closely related to EP2 that also couples to stimulatory guanine nucleotide binding protein, but the functional differences between EP2 and EP4 in Mtb‐infected Mφs have been unclear. EP4 antagonist addition to H37Ra‐infected Mφs inhibited the expression of cyclooxygenase 2 (COX2) and microsomal prostaglandin E synthase‐1 (mPGES‐1), which are involved in PGE2 production. Moreover, H37Ra infection induced PGE2 production through the Toll‐like receptor (TLR) 2/p38 mitogen‐activated protein kinase (MAPK) signaling pathway. Induction of COX2 and mPGES‐1 expression by TLR2 stimulation or Mtb infection was increased after additional stimulation with EP4 agonist. Hence, in Mtb‐infected Mφs, PGE2 production induced by pathogen recognition receptors/p38 MAPK signaling is upregulated by EP4‐triggered signaling to maintain an effective PGE2 concentration.—Nishimura, T., Zhao, X., Gan, H., Koyasu, S., and Remold, H. G., The prostaglandin E2 receptor EP4 is integral to a positive feedback loop for prostaglandin E2 production in human macrophages infected with Mycobacterium tuberculosis. FASEB J. 27, 3827‐3836 (2013). www.fasebj.org


Mucosal Immunology | 2017

Bcl-xL mediates RIPK3-dependent necrosis in M. tuberculosis-infected macrophages

Xiaomin Zhao; Nargis Khan; Huixian Gan; Fanny Tzelepis; Tomoyasu Nishimura; Seung-Yeol Park; Maziar Divangahi; Heinz G. Remold

Virulent Mycobacterium tuberculosis (Mtb) triggers necrosis in host Mϕ, which is essential for successful pathogenesis in tuberculosis. Here we demonstrate that necrosis of Mtb-infected Mϕ is dependent on the action of the cytosolic Receptor Interacting Protein Kinase 3 (RIPK3) and the mitochondrial Bcl-2 family member protein B-cell lymphoma—extra large (Bcl-xL). RIPK3-deficient Mϕ are able to better control bacterial growth in vitro and in vivo. Mechanistically, cytosolic RIPK3 translocates to the mitochondria where it promotes necrosis and blocks caspase 8-activation and apoptosis via Bcl-xL. Furthermore, necrosis is associated with stabilization of hexokinase II on the mitochondria as well as cyclophilin D-dependent mitochondrial permeability transition. Collectively, these events upregulate the level of reactive oxygen species to induce necrosis. Thus, in Mtb-infected Mϕ, mitochondria are an essential platform for induction of necrosis by activating RIPK3 function and preventing caspase 8-activation.

Collaboration


Dive into the Huixian Gan's collaboration.

Top Co-Authors

Avatar

Heinz G. Remold

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Minjian Chen

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Maziar Divangahi

McGill University Health Centre

View shared research outputs
Top Co-Authors

Avatar

Samuel M. Behar

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David M. Lee

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Fucheng Ren

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Hardy Kornfeld

Boston University Medical Campus

View shared research outputs
Researchain Logo
Decentralizing Knowledge