Network


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

Hotspot


Dive into the research topics where Trevor Clarke is active.

Publication


Featured researches published by Trevor Clarke.


Proceedings of the National Academy of Sciences of the United States of America | 2010

CXCR4 blockade augments bone marrow progenitor cell recruitment to the neovasculature and reduces mortality after myocardial infarction

Kentaro Jujo; Hiromichi Hamada; Atsushi Iwakura; Tina Thorne; Haruki Sekiguchi; Trevor Clarke; Aiko Ito; Sol Misener; Toshikazu Tanaka; Ekaterina Klyachko; Koichi Kobayashi; Jörn Tongers; Jérôme Roncalli; Yukio Tsurumi; Nobuhisa Hagiwara; Douglas W. Losordo

We hypothesized that a small molecule CXCR4 antagonist, AMD3100 (AMD), could augment the mobilization of bone marrow (BM)-derived endothelial progenitor cells (EPCs), thereby enhancing neovascularization and functional recovery after myocardial infarction. Single-dose AMD injection administered after the onset of myocardial infarction increased circulating EPC counts and myocardial vascularity, reduced fibrosis, and improved cardiac function and survival. In mice transplanted with traceable BM cells, AMD increased BM-derived cell incorporation in the ischemic border zone. In contrast, continuous infusion of AMD, although increasing EPCs in the circulation, worsened outcome by blocking EPC incorporation. In addition to its effects as a CXCR4 antagonist, AMD also up-regulated VEGF and matrix metalloproteinase 9 (MMP-9) expression, and the benefits of AMD were not observed in the absence of MMP-9 expression in the BM. These findings suggest that AMD3100 preserves cardiac function after myocardial infarction by enhancing BM-EPC–mediated neovascularization, and that these benefits require MMP-9 expression in the BM, but not in the ischemic region. Our results indicate that AMD3100 could be a potentially useful therapy for the treatment of myocardial infarction.


Circulation | 2013

CXC-Chemokine Receptor 4 Antagonist AMD3100 Promotes Cardiac Functional Recovery After Ischemia/Reperfusion Injury via Endothelial Nitric Oxide Synthase–Dependent Mechanism

Kentaro Jujo; Masaaki; Haruki Sekiguchi; Ekaterina Klyachko; Sol Misener; Toshikazu Tanaka; Jörn Tongers; Jérôme Roncalli; Marie Ange Renault; Tina Thorne; Aiko Ito; Trevor Clarke; Christine Kamide; Yukio Tsurumi; Nobuhisa Hagiwara; Gangjian Qin; Michio Asahi; Douglas W. Losordo

Background— CXC-chemokine receptor 4 (CXCR4) regulates the retention of stem/progenitor cells in the bone marrow (BM), and the CXCR4 antagonist AMD3100 improves recovery from coronary ligation injury by mobilizing stem/progenitor cells from the BM to the peripheral blood. Thus, we investigated whether AMD3100 also improves recovery from ischemia/reperfusion injury, which more closely mimics myocardial infarction in patients, because blood flow is only temporarily obstructed. Methods and Results— Mice were treated with single subcutaneous injections of AMD3100 (5 mg/kg) or saline after ischemia/reperfusion injury. Three days later, histological measurements of the ratio of infarct area to area at risk were smaller in AMD3100-treated mice than in mice administered saline, and echocardiographic measurements of left ventricular function were greater in the AMD3100-treated mice at week 4. CXCR4+ cells were mobilized for just 1 day in both groups, but the mobilization of sca1+/flk1+ cells endured for 7 days in AMD3100-treated mice compared with just 1 day in the saline-treated mice. AMD3100 upregulated BM levels of endothelial nitric oxide synthase (eNOS) and 2 targets of eNOS signaling, matrix metalloproteinase-9 and soluble Kit ligand. Furthermore, the loss of BM eNOS expression abolished the benefit of AMD3100 on sca1+/flk1+ cell mobilization without altering the mobilization of CXCR4+ cells, and the cardioprotective effects of AMD3100 were retained in eNOS-knockout mice that had been transplanted with BM from wild-type mice but not in wild-type mice with eNOS-knockout BM. Conclusions— AMD3100 prolongs BM progenitor mobilization and improves recovery from ischemia/reperfusion injury, and these benefits appear to occur through a previously unidentified link between AMD3100 and BM eNOS expression.


Circulation Research | 2009

The Hedgehog Transcription Factor Gli3 Modulates Angiogenesis

Marie-Ange Renault; Jérôme Roncalli; Jörn Tongers; Sol Misener; Tina Thorne; Kentaro Jujo; Aiko Ito; Trevor Clarke; Chris Fung; Meredith Millay; Christine Kamide; Andrew Scarpelli; Ekaterina Klyachko; Douglas W. Losordo

Rationale: The Gli transcription factors are mediators of Hedgehog (Hh) signaling and have been shown to play critical roles during embryogenesis. Previously, we have demonstrated that the Hh pathway is reactivated by ischemia in adult mammals, and that this pathway can be stimulated for therapeutic benefit; however, the specific roles of the Gli transcription factors during ischemia-induced Hh signaling have not been elucidated. Objective: To investigate the role of Gli3 in ischemic tissue repair. Methods and Results: Gli3-haploinsufficient (Gli3+/−) mice and their wild-type littermates were physiologically similar in the absence of ischemia; however, histological assessments of capillary density and echocardiographic measurements of left ventricular ejection fractions were reduced in Gli3+/− mice compared to wild-type mice after surgically induced myocardial infarction, and fibrosis was increased. Gli3-deficient mice also displayed reduced capillary density after induction of hindlimb ischemia and an impaired angiogenic response to vascular endothelial growth factor in the corneal angiogenesis model. In endothelial cells, adenovirus-mediated overexpression of Gli3 promoted migration (modified Boyden chamber), small interfering RNA–mediated downregulation of Gli3 delayed tube formation (Matrigel), and Western analyses identified increases in Akt phosphorylation, extracellular signal-regulated kinase (ERK)1/2 activation, and c-Fos expression; however, promoter–reporter assays indicated that Gli3 overexpression does not modulate Gli-dependent transcription. Furthermore, the induction of endothelial cell migration by Gli3 was dependent on Akt and ERK1/2 activation. Conclusions: Collectively, these observations indicate that Gli3 contributes to vessel growth under both ischemic and nonischemic conditions and provide the first evidence that Gli3 regulates angiogenesis and endothelial cell activity in adult mammals.


Laboratory Investigation | 2012

Estradiol triggers sonic-hedgehog-induced angiogenesis during peripheral nerve regeneration by downregulating hedgehog-interacting protein

Haruki Sekiguchi; Masaaki; Kentaro Jujo; Marie-Ange Renault; Tina Thorne; Trevor Clarke; Aiko Ito; Toshikazu Tanaka; Ekaterina Klyachko; Yasuhiko Tabata; Nobuhisa Hagiwara; Douglas W. Losordo

Both estradiol (E2) and Sonic Hedgehog (Shh) contribute to angiogenesis and nerve regeneration. Here, we investigated whether E2 improves the recovery of injured nerves by downregulating the Shh inhibitor hedgehog-interacting protein (HIP) and increasing Shh-induced angiogenesis. Mice were treated with local injections of E2 or placebo one week before nerve-crush injury; 28 days after injury, nerve conduction velocity, exercise duration, and vascularity were significantly greater in E2-treated mice than in placebo-treated mice. E2 treatment was also associated with higher mRNA levels of Shh, the Shh receptor Patched-1, and the Shh transcriptional target Gli1, but with lower levels of HIP. The E2-induced enhancement of nerve vascularity was abolished by the Shh inhibitor cyclopamine, and the effect of E2 treatment on Shh, Gli1, and HIP mRNA expression was abolished by the E2 inhibitor ICI. Gli-luciferase activity in human umbilical-vein endothelial cells (HUVECs) increased more after treatment with E2 and Shh than after treatment with E2 alone, and E2 treatment reduced HIP expression in HUVECs and Schwann cells without altering Shh expression. Collectively, these findings suggest that E2 improves nerve recovery, at least in part, by reducing HIP expression, which subsequently leads to an increase in Shh signaling and Shh-induced angiogenesis.


Circulation | 2012

CXCR4 Antagonist AMD3100 Promotes Cardiac Functional Recovery After Ischemia-Reperfusion Injury via eNOS-Dependent Mechanism

Kentaro Jujo; Masaaki; Haruki Sekiguchi; Ekaterina Klyachko; Sol Misener; Toshikazu Tanaka; Jörn Tongers; Jérôme Roncalli; Marie-Ange Renault; Tina Thorne; Aiko Ito; Trevor Clarke; Christine Kamide; Yukio Tsurumi; Nobuhisa Hagiwara; Gangjian Qin; Michio Asahi; Douglas W. Losordo

Background— CXC-chemokine receptor 4 (CXCR4) regulates the retention of stem/progenitor cells in the bone marrow (BM), and the CXCR4 antagonist AMD3100 improves recovery from coronary ligation injury by mobilizing stem/progenitor cells from the BM to the peripheral blood. Thus, we investigated whether AMD3100 also improves recovery from ischemia/reperfusion injury, which more closely mimics myocardial infarction in patients, because blood flow is only temporarily obstructed. Methods and Results— Mice were treated with single subcutaneous injections of AMD3100 (5 mg/kg) or saline after ischemia/reperfusion injury. Three days later, histological measurements of the ratio of infarct area to area at risk were smaller in AMD3100-treated mice than in mice administered saline, and echocardiographic measurements of left ventricular function were greater in the AMD3100-treated mice at week 4. CXCR4+ cells were mobilized for just 1 day in both groups, but the mobilization of sca1+/flk1+ cells endured for 7 days in AMD3100-treated mice compared with just 1 day in the saline-treated mice. AMD3100 upregulated BM levels of endothelial nitric oxide synthase (eNOS) and 2 targets of eNOS signaling, matrix metalloproteinase-9 and soluble Kit ligand. Furthermore, the loss of BM eNOS expression abolished the benefit of AMD3100 on sca1+/flk1+ cell mobilization without altering the mobilization of CXCR4+ cells, and the cardioprotective effects of AMD3100 were retained in eNOS-knockout mice that had been transplanted with BM from wild-type mice but not in wild-type mice with eNOS-knockout BM. Conclusions— AMD3100 prolongs BM progenitor mobilization and improves recovery from ischemia/reperfusion injury, and these benefits appear to occur through a previously unidentified link between AMD3100 and BM eNOS expression.


Circulation | 2013

CXC-Chemokine Receptor 4 Antagonist AMD3100 Promotes Cardiac Functional Recovery After Ischemia/Reperfusion Injury via Endothelial Nitric Oxide Synthase–Dependent MechanismClinical Perspective

Kentaro Jujo; Masaaki; Haruki Sekiguchi; Ekaterina Klyachko; Sol Misener; Toshikazu Tanaka; Jörn Tongers; Jérôme Roncalli; Marie-Ange Renault; Tina Thorne; Aiko Ito; Trevor Clarke; Christine Kamide; Yukio Tsurumi; Nobuhisa Hagiwara; Gangjian Qin; Michio Asahi; Douglas W. Losordo

Background— CXC-chemokine receptor 4 (CXCR4) regulates the retention of stem/progenitor cells in the bone marrow (BM), and the CXCR4 antagonist AMD3100 improves recovery from coronary ligation injury by mobilizing stem/progenitor cells from the BM to the peripheral blood. Thus, we investigated whether AMD3100 also improves recovery from ischemia/reperfusion injury, which more closely mimics myocardial infarction in patients, because blood flow is only temporarily obstructed. Methods and Results— Mice were treated with single subcutaneous injections of AMD3100 (5 mg/kg) or saline after ischemia/reperfusion injury. Three days later, histological measurements of the ratio of infarct area to area at risk were smaller in AMD3100-treated mice than in mice administered saline, and echocardiographic measurements of left ventricular function were greater in the AMD3100-treated mice at week 4. CXCR4+ cells were mobilized for just 1 day in both groups, but the mobilization of sca1+/flk1+ cells endured for 7 days in AMD3100-treated mice compared with just 1 day in the saline-treated mice. AMD3100 upregulated BM levels of endothelial nitric oxide synthase (eNOS) and 2 targets of eNOS signaling, matrix metalloproteinase-9 and soluble Kit ligand. Furthermore, the loss of BM eNOS expression abolished the benefit of AMD3100 on sca1+/flk1+ cell mobilization without altering the mobilization of CXCR4+ cells, and the cardioprotective effects of AMD3100 were retained in eNOS-knockout mice that had been transplanted with BM from wild-type mice but not in wild-type mice with eNOS-knockout BM. Conclusions— AMD3100 prolongs BM progenitor mobilization and improves recovery from ischemia/reperfusion injury, and these benefits appear to occur through a previously unidentified link between AMD3100 and BM eNOS expression.


Circulation | 2008

Abstract 5445: Gli2 and Gli3 Are Over-expressed in the Ischemic Tissue and Participate in Ischemia-induced Angiogenesis and Myogenesis

Marie-Ange Renault; Jérôme Roncalli; Joern Tongers; Hiromichi Hamada; Tina Thorne; Sol Misener; Aiko Ito; Trevor Clarke; Meredith Millay; Andrew Scarpelli; Ekaterina Klyachko; Douglas W. Losordo


Archive | 2012

Ischemia-Reperfusion Injury via eNOS-Dependent Mechanism CXCR4 Antagonist AMD3100 Promotes Cardiac Functional Recovery After

Yukio Tsurumi; Nobuhisa Hagiwara; Gangjian Qin; Michio Asahi; Douglas W. Losordo; Jörn Tongers; Jérôme Roncalli; Marie-Ange Renault; Aiko Ito; Trevor Clarke; Kentaro Jujo; Masaaki; Haruki Sekiguchi; Ekaterina Klyachko; Sol Misener; Toshikazu Tanaka


Circulation | 2009

Abstract 5635: Estradiol Promotes Neural Stem Cell-based Peripheral Nerve Repair by Enhancing Angiogenesis

Haruki Sekiguchi; Masaaki; Kentaro Jujo; Tina Thorne; Andy Scarpelli; Trevor Clarke; Aiko Ito; Sol Misener; Ekaterina Klyachko; Nobuhisa Hagiwara; Douglas W. Losordo


Circulation | 2009

Abstract 3701: CXCR4 Antagonist AMD3100 Preserves Cardiac Function After Ischemia-Reperfusion Injury via eNOS Dependent Mobilization of Bone Marrow Derived Progenitor Cells

Kentaro Jujo; Ekaterina Klyachko; Sol Misener; Tina Thorne; Masaaki; Haruki Sekiguchi; Toshikazu Tanaka; Joern Tongers; Jérôme Roncalli; Trevor Clarke; Aiko Ito; Christine Kamide; Yukio Tsurumi; Nobuhisa Hagiwara; Gangjian Qin; Douglas W. Losordo

Collaboration


Dive into the Trevor Clarke's collaboration.

Top Co-Authors

Avatar

Aiko Ito

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tina Thorne

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Kentaro Jujo

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Sol Misener

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge