Network


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

Hotspot


Dive into the research topics where Enming J. Su is active.

Publication


Featured researches published by Enming J. Su.


Nature Medicine | 2008

Activation of PDGF-CC by tissue plasminogen activator impairs blood-brain barrier integrity during ischemic stroke

Enming J. Su; Linda Fredriksson; Melissa Geyer; Erika Folestad; Jacqueline M. Cale; Johanna Andrae; Yamei Gao; Kristian Pietras; Kris Mann; Manuel Yepes; Dudley K. Strickland; Christer Betsholtz; Ulf Eriksson; Daniel A. Lawrence

Thrombolytic treatment of ischemic stroke with tissue plasminogen activator (tPA) is markedly limited owing to concerns about hemorrhagic complications and the requirement that tPA be administered within 3 h of symptoms. Here we report that tPA activation of latent platelet-derived growth factor-CC (PDGF-CC) may explain these limitations. Intraventricular injection of tPA or active PDGF-CC, in the absence of ischemia, leads to significant increases in cerebrovascular permeability. In contrast, co-injection of neutralizing antibodies to PDGF-CC with tPA blocks this increased permeability, indicating that PDGF-CC is a downstream substrate of tPA within the neurovascular unit. These effects are mediated through activation of PDGF-α receptors (PDGFR-α) on perivascular astrocytes, and treatment of mice with the PDGFR-α antagonist imatinib after ischemic stroke reduces both cerebrovascular permeability and hemorrhagic complications associated with late administration of thrombolytic tPA. These data demonstrate that PDGF signaling regulates blood-brain barrier permeability and suggest potential new strategies for stroke treatment.


The EMBO Journal | 2006

Endocytic receptor LRP together with tPA and PAI-1 coordinates Mac-1-dependent macrophage migration.

Chunzhang Cao; Daniel A. Lawrence; Yang Li; Joachim Herz; Enming J. Su; Alexandra Makarova; Bradley T. Hyman; Dudley K. Strickland; Li Zhang

Migration of activated macrophages is essential for resolution of acute inflammation and the initiation of adaptive immunity. Here, we show that efficient macrophage migration in inflammatory environment depends on Mac‐1 recognition of a binary complex consisting of fibrin within the provisional matrix and the protease tPA (tissue‐type plasminogen activator). Subsequent neutralization of tPA by its inhibitor PAI‐1 enhances binding of the integrin–protease–inhibitor complex to the endocytic receptor LRP (lipoprotein receptor‐related protein), triggering a switch from cell adhesion to cell detachment. Genetic inactivation of Mac‐1, tPA, PAI‐1 or LRP but not the protease uPA abrogates macrophage migration. The defective macrophage migration in PAI‐1‐deficient mice can be restored by wild‐type but not by a mutant PAI‐1 that does not interact with LRP. In vitro analysis shows that tPA promotes Mac‐1‐mediated adhesion, whereas PAI‐1 and LRP facilitate its transition to cell retraction. Our results emphasize the importance of ordered transitions both temporally and spatially between individual steps of cell migration, and support a model where efficient migration of inflammatory macrophages depends on cooperation of three physiologically prominent systems (integrins, coagulation and fibrinolysis, and endocytosis).


Journal of Clinical Investigation | 2009

Self-regulation of inflammatory cell trafficking in mice by the leukocyte surface apyrase CD39

Matthew C. Hyman; Danica Petrovic-Djergovic; Scott H. Visovatti; Hui Liao; Sunitha Yanamadala; Diane Bouis; Enming J. Su; Daniel A. Lawrence; M. Johan Broekman; Aaron J. Marcus; David J. Pinsky

Leukocyte and platelet accumulation at sites of cerebral ischemia exacerbate cerebral damage. The ectoenzyme CD39 on the plasmalemma of endothelial cells metabolizes ADP to suppress platelet accumulation in the ischemic brain. However, the role of leukocyte surface CD39 in regulating monocyte and neutrophil trafficking in this setting is not known. Here we have demonstrated in mice what we believe to be a novel mechanism by which CD39 on monocytes and neutrophils regulates their own sequestration into ischemic cerebral tissue, by catabolizing nucleotides released by injured cells, thereby inhibiting their chemotaxis, adhesion, and transmigration. Bone marrow reconstitution and provision of an apyrase, an enzyme that hydrolyzes nucleoside tri- and diphosphates, each normalized ischemic leukosequestration and cerebral infarction in CD39-deficient mice. Leukocytes purified from Cd39-/- mice had a markedly diminished capacity to phosphohydrolyze adenine nucleotides and regulate platelet reactivity, suggesting that leukocyte ectoapyrases modulate the ambient vascular nucleotide milieu. Dissipation of ATP by CD39 reduced P2X7 receptor stimulation and thereby suppressed baseline leukocyte alphaMbeta2-integrin expression. As alphaMbeta2-integrin blockade reversed the postischemic, inflammatory phenotype of Cd39-/- mice, these data suggest that phosphohydrolytic activity on the leukocyte surface suppresses cell-cell interactions that would otherwise promote thrombosis or inflammation. These studies indicate that CD39 on both endothelial cells and leukocytes reduces inflammatory cell trafficking and platelet reactivity, with a consequent reduction in tissue injury following cerebral ischemic challenge.


Journal of Thrombosis and Haemostasis | 2006

Effect of pharmacologic plasminogen activator inhibitor‐1 inhibition on cell motility and tumor angiogenesis

Courtney E. Leik; Enming J. Su; P. Nambi; David L. Crandall; Daniel A. Lawrence

Summary.  Background: Plasminogen activator inhibitor‐1 (PAI‐1) is integrally involved in tumorigenesis by impacting on both proteolytic activity and cell migration during angiogenesis. Objectives: We hypothesized that an orally active small molecule inhibitor of PAI‐1 (PAI‐039; tiplaxtinin) could affect smooth muscle cell (SMC) attachment and migration in vitro on a vitronectin matrix, and exhibit antiangiogenic activity in vivo. Methods: In vitro assays were used to assess the mechanism of inhibition of PAI‐1 by PAI‐039 using wild‐type PAI‐1 in the presence or absence of vitronectin and wild‐type PAI‐1 and specific PAI‐1 mutants in SMC adhesion and migration assays. An in vivo tumor angiogenesis model was used to assess the effect of PAI‐039 administration on neovascularization in a Matrigel implant. Results: PAI‐039 dose‐dependently inhibited soluble, but not vitronectin‐bound, PAI‐1. Cell adhesion assays using PAI‐1 mutants unable to bind vitronectin (PAI‐1K) or inactivate proteases (PAI‐1R) further suggested that PAI‐039 inactivated PAI‐1 by binding near its vitronectin domain. In a tumor angiogenesis model, PAI‐039 treatment of wild‐type mice dose‐dependently decreased hemoglobin concentration and endothelial cell staining within the Matrigel implant, indicating reduced angiogenesis, but exhibited no in vivo efficacy in PAI‐1 null mice. Conclusions: Administration of an orally active PAI‐1 inhibitor prevented angiogenesis in a Matrigel implant. The lack of activity of PAI‐039 against wild‐type PAI‐1 bound to vitronectin and PAI‐1K suggests PAI‐039 binding near the vitronectin‐binding site. Our studies further substantiate a role for PAI‐1 in cellular motility and tumor angiogenesis, and suggest for the first time that these effects can be modulated pharmacologically.


Brain | 2012

The tissue-type plasminogen activator–plasminogen activator inhibitor 1 complex promotes neurovascular injury in brain trauma: evidence from mice and humans

Maithili Sashindranath; Eunice Sales; Maria Daglas; Roxann Freeman; Andre L. Samson; Elisa J. Cops; Simone A. Beckham; Adam Galle; Catriona McLean; Cristina Morganti-Kossmann; Jeffrey V. Rosenfeld; Rime Madani; Jean-Dominique Vassalli; Enming J. Su; Daniel A. Lawrence; Robert L. Medcalf

The neurovascular unit provides a dynamic interface between the circulation and central nervous system. Disruption of neurovascular integrity occurs in numerous brain pathologies including neurotrauma and ischaemic stroke. Tissue plasminogen activator is a serine protease that converts plasminogen to plasmin, a protease that dissolves blood clots. Besides its role in fibrinolysis, tissue plasminogen activator is abundantly expressed in the brain where it mediates extracellular proteolysis. However, proteolytically active tissue plasminogen activator also promotes neurovascular disruption after ischaemic stroke; the molecular mechanisms of this process are still unclear. Tissue plasminogen activator is naturally inhibited by serine protease inhibitors (serpins): plasminogen activator inhibitor-1, neuroserpin or protease nexin-1 that results in the formation of serpin:protease complexes. Proteases and serpin:protease complexes are cleared through high-affinity binding to low-density lipoprotein receptors, but their binding to these receptors can also transmit extracellular signals across the plasma membrane. The matrix metalloproteinases are the second major proteolytic system in the mammalian brain, and like tissue plasminogen activators are pivotal to neurological function but can also degrade structures of the neurovascular unit after injury. Herein, we show that tissue plasminogen activator potentiates neurovascular damage in a dose-dependent manner in a mouse model of neurotrauma. Surprisingly, inhibition of activity following administration of plasminogen activator inhibitor-1 significantly increased cerebrovascular permeability. This led to our finding that formation of complexes between tissue plasminogen activator and plasminogen activator inhibitor-1 in the brain parenchyma facilitates post-traumatic cerebrovascular damage. We demonstrate that following trauma, the complex binds to low-density lipoprotein receptors, triggering the induction of matrix metalloproteinase-3. Accordingly, pharmacological inhibition of matrix metalloproteinase-3 attenuates neurovascular permeability and improves neurological function in injured mice. Our results are clinically relevant, because concentrations of tissue plasminogen activator: plasminogen activator inhibitor-1 complex and matrix metalloproteinase-3 are significantly elevated in cerebrospinal fluid of trauma patients and correlate with neurological outcome. In a separate study, we found that matrix metalloproteinase-3 and albumin, a marker of cerebrovascular damage, were significantly increased in brain tissue of patients with neurotrauma. Perturbation of neurovascular homeostasis causing oedema, inflammation and cell death is an important cause of acute and long-term neurological dysfunction after trauma. A role for the tissue plasminogen activator-matrix metalloproteinase axis in promoting neurovascular disruption after neurotrauma has not been described thus far. Targeting tissue plasminogen activator: plasminogen activator inhibitor-1 complex signalling or downstream matrix metalloproteinase-3 induction may provide viable therapeutic strategies to reduce cerebrovascular permeability after neurotrauma.


Journal of Biological Chemistry | 2007

The contributions of integrin affinity and integrin-cytoskeletal engagement in endothelial and smooth muscle cell adhesion to vitronectin

Steingrimur Stefansson; Enming J. Su; Shoji Ishigami; Jacqueline M. Cale; Yamei Gao; Natalia V. Gorlatova; Daniel A. Lawrence

The serine proteinase inhibitor, plasminogen activator inhibitor type-1 (PAI-1), binds to the adhesion protein vitronectin with high affinity at a site that is located directly adjacent to the vitronectin RGD integrin binding sequence. The binding of PAI-1 to vitronectin sterically blocks integrin access to this site and completely inhibits the binding of purified integrins to vitronectin; however, its inhibition of endothelial and smooth muscle cell adhesion to vitronectin is at most 50-75%. Because PAI-1 binds vitronectin with ∼10-100-fold higher affinity than purified integrins, we have analyzed the mechanism whereby these cells are able to overcome this obstacle. Our studies exclude proteolytic removal of PAI-1 from vitronectin as the mechanism, and show instead that cell adhesion in the presence of PAI-1 is dependent on integrin-cytoskeleton engagement. Disrupting endothelial or smooth muscle cell actin polymerization and/or focal adhesion assembly reduces cell adhesion to vitronectin in the presence of PAI-1 to levels similar to that observed for the binding of purified integrins to vitronectin. Furthermore, endothelial cell, but not smooth muscle cell adhesion to vitronectin in the presence of PAI-1 requires both polymerized microtubules and actin, further demonstrating the importance of the cytoskeleton for integrin-mediated adhesion. Finally, we show that cell adhesion in the presence of PAI-1 leads to colocalization of PAI-1 with the integrins αvβ3 and αvβ5 at the cell-matrix interface.


Journal of Biological Chemistry | 2010

Characterization of a Novel Class of Polyphenolic Inhibitors of Plasminogen Activator Inhibitor-1

Jacqueline M. Cale; Shih Hon Li; Mark Warnock; Enming J. Su; Paul R. North; Karen L. Sanders; Maria M. Puscau; Cory D. Emal; Daniel A. Lawrence

Plasminogen activator inhibitor type 1, (PAI-1) the primary inhibitor of the tissue-type (tPA) and urokinase-type (uPA) plasminogen activators, has been implicated in a wide range of pathological processes, making it an attractive target for pharmacologic inhibition. Currently available small-molecule inhibitors of PAI-1 bind with relatively low affinity and do not inactivate PAI-1 in the presence of its cofactor, vitronectin. To search for novel PAI-1 inhibitors with improved potencies and new mechanisms of action, we screened a library selected to provide a range of biological activities and structural diversity. Five potential PAI-1 inhibitors were identified, and all were polyphenolic compounds including two related, naturally occurring plant polyphenols that were structurally similar to compounds previously shown to provide cardiovascular benefit in vivo. Unique second generation compounds were synthesized and characterized, and several showed IC50 values for PAI-1 between 10 and 200 nm. This represents an enhanced potency of 10–1000-fold over previously reported PAI-1 inactivators. Inhibition of PAI-1 by these compounds was reversible, and their primary mechanism of action was to block the initial association of PAI-1 with a protease. Consistent with this mechanism and in contrast to previously described PAI-1 inactivators, these compounds inactivate PAI-1 in the presence of vitronectin. Two of the compounds showed efficacy in ex vivo plasma and one blocked PAI-1 activity in vivo in mice. These data describe a novel family of high affinity PAI-1-inactivating compounds with improved characteristics and in vivo efficacy, and suggest that the known cardiovascular benefits of dietary polyphenols may derive in part from their inactivation of PAI-1.


Journal of Thrombosis and Haemostasis | 2009

Tissue plasminogen activator-mediated PDGF signaling and neurovascular coupling in stroke

Enming J. Su; Linda Fredriksson; Gerald P. Schielke; Ulf Eriksson; Daniel E. Lawrence

Summary.  The use of tissue plasminogen activator (tPA) as a thrombolytic treatment in ischemic stroke is limited largely due to concerns for hemorrhagic complications. The underlying mechanisms are still unknown, but evidence is beginning to emerge that tPA interacts with key regulators of the neurovascular unit (NVU), and that these interactions may contribute to the undesirable side effects associated with the use of tPA in ischemic stroke. Understanding these connections and tPA’s normal function within the NVU may offer new insights into future therapeutic approaches.


American Journal of Pathology | 2012

Platelet-Derived Growth Factor C Deficiency in C57BL/6 Mice Leads to Abnormal Cerebral Vascularization, Loss of Neuroependymal Integrity, and Ventricular Abnormalities

Linda Fredriksson; Ingrid Nilsson; Enming J. Su; Johanna Andrae; Hao Ding; Christer Betsholtz; Ulf Eriksson; Daniel A. Lawrence

Platelet-derived growth factors (PDGFs) and their tyrosine kinase receptors (PDGFRs) are known to play important roles during development of the lungs, central nervous system (CNS), and skeleton and in several diseases. PDGF-C is a ligand for the tyrosine kinase receptor PDGFRα. Mutations in the gene encoding PDGF-C have been linked to clefts of the lip and/or palate in humans, and ablation of PDGF-C in 129/Sv background mice results in death during the perinatal period. In this study, we report that ablation of PDGF-C in C57BL/6 mice results in a milder phenotype than in 129/Sv mice, and we present a phenotypic characterization of PDGF-C deficiency in the adult murine CNS. Multiple congenital defects were observed in the CNS of PDGF-C-null C57BL/6 mice, including cerebral vascular abnormalities with abnormal vascular smooth muscle cell coverage. In vivo imaging of mice deficient in PDGF-C also revealed cerebral ventricular abnormalities, such as asymmetry of the lateral ventricles and hypoplasia of the septum, reminiscent of cavum septum pellucidum in humans. We further noted that PDGF-C-deficient mice displayed a distorted ependymal lining of the lateral ventricles, and we found evidence of misplaced neurons in the ventricular lining. We conclude that PDGF-C plays a critical role in the development of normal cerebral ventricles and neuroependymal integrity as well as in normal cerebral vascularization.


Journal of Thrombosis and Haemostasis | 2011

The thrombomodulin analog Solulin promotes reperfusion and reduces infarct volume in a thrombotic stroke model

Enming J. Su; M. Geyer; M. Wahl; K. Mann; David Ginsburg; H. Brohmann; K. U. Petersen; Daniel A. Lawrence

See also Andreou AP, Crawley JTB. Thrombomodulin analogues for the treatment of ischemic stroke. This issue, pp 1171–3.

Collaboration


Dive into the Enming J. Su's collaboration.

Top Co-Authors

Avatar

Daniel A. Lawrence

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yamei Gao

University of Maryland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Diane Bouis

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge