Network


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

Hotspot


Dive into the research topics where Baikun Wang is active.

Publication


Featured researches published by Baikun Wang.


Immunity | 2002

Cytotoxic cell granule-mediated apoptosis: Perforin delivers granzyme b-serglycin complexes into target cells without plasma membrane pore formation

Sunil S. Metkar; Baikun Wang; Miguel Aguilar-Santelises; Srikumar M. Raja; Lars Uhlin-Hansen; Eckhard R. Podack; Joseph A. Trapani; Christopher J. Froelich

The mechanism underlying perforin (PFN)-dependent delivery of apoptotic granzymes during cytotoxic cell granule-mediated death remains speculative. Granzyme B (GrB) and perforin were found to coexist as multimeric complexes with the proteoglycan serglycin (SG) in cytotoxic granules, and cytotoxic cells were observed to secrete exclusively macromolecular GrB-SG. Contrary to the view that PFN acts as a gateway for granzymes through the plasma membrane, monomeric PFN and, strikingly, PFN-SG complexes were shown to mediate cytosolic delivery of macromolecular GrB-SG without producing detectable plasma membrane pores. These results indicate that granule-mediated apoptosis represents a phenomenon whereby the target cell perceives granule contents as a multimeric complex consisting of SG, PFN, and granzymes, which are, respectively, the scaffold, translocator, and targeting/informational components of this modular delivery system.


Immunity | 2008

Human and Mouse Granzyme A Induce a Proinflammatory Cytokine Response

Sunil S. Metkar; Cheikh Menaa; Julián Pardo; Baikun Wang; Reinhard Wallich; Marina A. Freudenberg; Stephen Kim; Srikumar M. Raja; Lianfa Shi; Markus M. Simon; Christopher J. Froelich

Granzyme A (GzmA) is considered a major proapoptotic protease. We have discovered that GzmA-induced cell death involves rapid membrane damage that depends on the synergy between micromolar concentrations of GzmA and sublytic perforin (PFN). Ironically, GzmA and GzmB, independent of their catalytic activity, both mediated this swift necrosis. Even without PFN, lower concentrations of human GzmA stimulated monocytic cells to secrete proinflammatory cytokines (interleukin-1beta [IL-1beta], TNFalpha, and IL-6) that were blocked by a caspase-1 inhibitor. Moreover, murine GzmA and GzmA(+) cytotoxic T lymphocytes (CTLs) induce IL-1beta from primary mouse macrophages, and GzmA(-/-) mice resist lipopolysaccharide-induced toxicity. Thus, the granule secretory pathway plays an unexpected role in inflammation, with GzmA acting as an endogenous modulator.


Journal of Cell Biology | 2003

Granzyme B activates procaspase-3 which signals a mitochondrial amplification loop for maximal apoptosis

Sunil S. Metkar; Baikun Wang; Michelle L. Ebbs; Jin H. Kim; Yong J. Lee; Srikumar M. Raja; Christopher J. Froelich

Granzyme B (GrB), acting similar to an apical caspase, efficiently activates a proteolytic cascade after intracellular delivery by perforin. Studies here were designed to learn whether the physiologic effector, GrB–serglycin, initiates apoptosis primarily through caspase-3 or through BH3-only proteins with subsequent mitochondrial permeabilization and apoptosis. Using four separate cell lines that were either genetically lacking the zymogen or rendered deficient in active caspase-3, we measured apoptotic indices within whole cells (active caspase-3, mitochondrial depolarization [ΔΨm] and TUNEL). Adhering to these conditions, the following were observed in targets after GrB delivery: (a) procaspase-3–deficient cells fail to display a reduced ΔΨm and DNA fragmentation; (b) Bax/Bak is required for optimal ΔΨm reduction, caspase-3 activation, and DNA fragmentation, whereas BID cleavage is undetected by immunoblot; (c) Bcl-2 inhibits GrB-mediated apoptosis (reduced ΔΨm and TUNEL reactivity) by blocking oligomerization of caspase-3; and (d) in procaspase-3–deficient cells a mitochondrial-independent pathway was identified which involved procaspase-7 activation, PARP cleavage, and nuclear condensation. The data therefore support the existence of a fully implemented apoptotic pathway initiated by GrB, propagated by caspase-3, and perpetuated by a mitochondrial amplification loop but also emphasize the presence of an ancillary caspase-dependent, mitochondria-independent pathway.


Journal of Biological Chemistry | 2002

Cytotoxic Cell Granule-mediated Apoptosis CHARACTERIZATION OF THE MACROMOLECULAR COMPLEX OF GRANZYME B WITH SERGLYCIN

Srikumar M. Raja; Baikun Wang; Mandakini Dantuluri; Umesh R. Desai; Borries Demeler; Katharina Spiegel; Sunil S. Metkar; Christopher J. Froelich

We have recently shown that the physiological mediator of granule-mediated apoptosis is a macromolecular complex of granzymes and perforin complexed with the chondroitin-sulfate proteoglycan, serglycin (Metkar, S. S., Wang, B., Aguilar-Santelises, M., Raja, S. M., Uhlin-Hansen, L., Podack, E., Trapani, J. A., and Froelich, C. J. (2002)Immunity 16, 417–428). We now report our biophysical studies establishing the nature of granzyme B-serglycin (GrB·SG) complex. Dynamic laser light scattering studies establish that SG has a hydrodynamic radius of ∼140 ± 23 nm, comparable to some viral particles. Agarose mobility shift gels and surface plasmon resonance (SPR), show that SG binds tightly to GrB and has the capacity to hold 30–60 GrB molecules. SPR studies also indicate equivalent binding affinities (K d ∼ 0.8 μm), under acidic (granule pH) and neutral isotonic conditions (extra-cytoplasmic pH), for GrB·SG interaction. Finally, characterization of GrB·SG interactions within granules revealed complexes of two distinct molecular sizes, one held ∼4–8 molecules of GrB, whereas the other contained as many as 32 molecules of GrB or other granule proteins. These studies provide a firm biophysical basis for our earlier reported observations that the proapoptotic granzyme is exocytosed predominantly as a macromolecular complex with SG.


PLOS ONE | 2011

Perforin Rapidly Induces Plasma Membrane Phospholipid Flip-Flop

Sunil S. Metkar; Baikun Wang; Elena Catalán; Gregor Anderluh; Robert J. C. Gilbert; Julián Pardo; Christopher J. Froelich

The cytotoxic cell granule secretory pathway is essential for host defense. This pathway is fundamentally a form of intracellular protein delivery where granule proteases (granzymes) from cytotoxic lymphocytes are thought to diffuse through barrel stave pores generated in the plasma membrane of the target cell by the pore forming protein perforin (PFN) and mediate apoptotic as well as additional biological effects. While recent electron microscopy and structural analyses indicate that recombinant PFN oligomerizes to form pores containing 20 monomers (20 nm) when applied to liposomal membranes, these pores are not observed by propidium iodide uptake in target cells. Instead, concentrations of human PFN that encourage granzyme-mediated apoptosis are associated with pore structures that unexpectedly favor phosphatidylserine flip-flop measured by Annexin-V and Lactadherin. Efforts that reduce PFN mediated Ca influx in targets did not reduce Annexin-V reactivity. Antigen specific mouse CD8 cells initiate a similar rapid flip-flop in target cells. A lipid that augments plasma membrane curvature as well as cholesterol depletion in target cells enhance flip-flop. Annexin-V staining highly correlated with apoptosis after Granzyme B (GzmB) treatment. We propose the structures that PFN oligomers form in the membrane bilayer may include arcs previously observed by electron microscopy and that these unusual structures represent an incomplete mixture of plasma membrane lipid and PFN oligomers that may act as a flexible gateway for GzmB to translocate across the bilayer to the cytosolic leaflet of target cells.


Cell Death & Differentiation | 2015

Perforin oligomers form arcs in cellular membranes: a locus for intracellular delivery of granzymes

Sunil S. Metkar; M Marchioretto; V. Antonini; L. Lunelli; Baikun Wang; R Jc Gilbert; Gregor Anderluh; R Roth; M Pooga; Julián Pardo; J E Heuser; Mauro Dalla Serra; Christopher J. Froelich

Perforin-mediated cytotoxicity is an essential host defense, in which defects contribute to tumor development and pathogenic disorders including autoimmunity and autoinflammation. How perforin (PFN) facilitates intracellular delivery of pro-apoptotic and inflammatory granzymes across the bilayer of targets remains unresolved. Here we show that cellular susceptibility to granzyme B (GzmB) correlates with rapid PFN-induced phosphatidylserine externalization, suggesting that pores are formed at a protein-lipid interface by incomplete membrane oligomers (or arcs). Supporting a role for these oligomers in protease delivery, an anti-PFN antibody (pf-80) suppresses necrosis but increases phosphatidylserine flip-flop and GzmB-induced apoptosis. As shown by atomic force microscopy on planar bilayers and deep-etch electron microscopy on mammalian cells, pf-80 increases the proportion of arcs which correlates with the presence of smaller electrical conductances, while large cylindrical pores decline. PFN appears to form arc structures on target membranes that serve as minimally disrupting conduits for GzmB translocation. The role of these arcs in PFN-mediated pathology warrants evaluation where they may serve as novel therapeutic targets.


Biochemical and Biophysical Research Communications | 2008

Granzyme B translocates across the lipid membrane only in the presence of lytic agents.

Mojca Podlesnik Beseničar; Sunil S. Metkar; Baikun Wang; Christopher J. Froelich; Gregor Anderluh

Granzyme B (GrB), a component of the cytotoxic cell granule secretion pathway, is designed to kill infected and transformed cells after intracellular delivery by the pore forming protein, perforin. The mechanism of the delivery remains speculative. In this study we tested the hypothesis that GrB possesses capacity to bind and disrupt lipid membranes. Here in comparison to previous studies that show GrB interacts with carbohydrate moieties, the protease does not bind membrane phospholipids nor has intrinsic membranolytic properties. To study the transmembrane movement of GrB, we developed a model membrane system consisting of a high-molecular weight GrB substrate encapsulated in unilamellar vesicles. Intra-vesicle proteolysis clearly requires concentrations of lytic agents (streptolysin O, perforin or Triton X-100) that disrupt unilamellar membranes.


Journal of Biological Chemistry | 1998

Granzyme B mimics apical caspases. Description of a unified pathway for trans-activation of executioner caspase-3 and -7.

Xiaohe Yang; Henning R. Stennicke; Baikun Wang; D R Green; Reiner U. Jänicke; Anu Srinivasan; Prem Seth; Guy S. Salvesen; Christopher J. Froelich


Journal of Biological Chemistry | 2005

A novel mechanism for protein delivery: Granzyme B undergoes electrostatic exchange from Serglycin to target cells

Srikumar M. Raja; Sunil S. Metkar; Stefan Höning; Baikun Wang; William A. Russin; Nina H. Pipalia; Cheikh Menaa; Mattias Belting; Xuefang Cao; Ralf Dressel; Christopher J. Froelich


Journal of Immunology | 1999

Apoptosis Induced by Granzyme B-Glycosaminoglycan Complexes: Implications for Granule-Mediated Apoptosis In Vivo

J. P. Galvin; L. H. A. Spaeny-Dekking; Baikun Wang; Prem Seth; C. E. Hack; Christopher J. Froelich

Collaboration


Dive into the Baikun Wang's collaboration.

Top Co-Authors

Avatar

Christopher J. Froelich

NorthShore University HealthSystem

View shared research outputs
Top Co-Authors

Avatar

Sunil S. Metkar

NorthShore University HealthSystem

View shared research outputs
Top Co-Authors

Avatar

Srikumar M. Raja

NorthShore University HealthSystem

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Borries Demeler

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Cheikh Menaa

NorthShore University HealthSystem

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mandakini Dantuluri

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Prem Seth

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge