Network


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

Hotspot


Dive into the research topics where Geoffrey Y. Akita is active.

Publication


Featured researches published by Geoffrey Y. Akita.


Circulation | 2000

Angiogenesis Is Induced in a Rabbit Model of Hindlimb Ischemia by Naked DNA Encoding an HIF-1α/VP16 Hybrid Transcription Factor

Karen A. Vincent; Kou-Gi Shyu; Yuxia Luo; Meredith Magner; Rene A. Tio; Canwen Jiang; Mark A. Goldberg; Geoffrey Y. Akita; Richard J. Gregory; Jeffrey M. Isner

BackgroundHypoxia-inducible factor-1 (HIF-1) is a heterodimeric transcription factor that regulates expression of genes involved in O2 homeostasis, including vascular endothelial growth factor (VEGF), a potent stimulator of angiogenesis. We sought to exploit this native adaptive response to hypoxia as a treatment for chronic ischemia. Methods and ResultsA hybrid protein consisting of DNA-binding and dimerization domains from the HIF-1&agr; subunit and the transactivation domain from herpes simplex virus VP16 protein was constructed to create a strong, constitutive transcriptional activator. After transfection into HeLa, C6, and Hep3B cells, this chimeric transcription factor was shown to activate expression of the endogenous VEGF gene, as well as several other HIF-1 target genes in vitro. The bioactivity of HIF-1&agr;/VP16 hybrid gene transfer in vivo was examined in a rabbit model of hindlimb ischemia. Administration of HIF-1&agr;/VP16 was associated with significant improvements in calf blood pressure ratio, angiographic score, resting and maximal regional blood flow, and capillary density (all P <0.01). ConclusionsThe HIF-1&agr;/VP16 hybrid transcription factor is able to promote significant improvement in perfusion of an ischemic limb. These results confirm the feasibility of a novel approach for therapeutic angiogenesis in which neovascularization may be achieved indirectly by use of a transcriptional regulatory strategy.


Circulation Research | 2003

Hypoxia-Inducible Factor-1 Mediates Activation of Cultured Vascular Endothelial Cells by Inducing Multiple Angiogenic Factors

Midori Yamakawa; Louis X. Liu; Taro Date; Adam J. Belanger; Karen A. Vincent; Geoffrey Y. Akita; Takayuki Kuriyama; Seng H. Cheng; Richard J. Gregory; Canwen Jiang

Abstract— Hypoxia-inducible factor-1 (HIF-1) mediates transcriptional activation of vascular endothelial growth factor (VEGF) and other hypoxia-responsive genes. Transgenic expression of a constitutively stable HIF-1&agr; mutant increases the number of vascular vessels without vascular leakage, tissue edema, or inflammation. This study aimed to investigate the molecular basis by which HIF-1 mediates the angiogenic response to hypoxia. In primary human endothelial cells, hypoxia, desferrioxamine, or infection with Ad2/HIF-1&agr;/VP16, an adenoviral vector encoding a constitutively stable hybrid form of HIF-1&agr;, increased the mRNA and protein levels of VEGF, angiopoietin-2 (Ang-2), and angiopoietin-4 (Ang-4). Infection with Ad2/CMVEV (a control vector expressing no transgene) had no effect. Angiopoietin-1 (Ang-1) expression was not detected in human endothelial cells. Ang-4 was also induced by hypoxia or Ad2/HIF-1&agr;/VP16 in human cardiac cells, whereas Ang-1 expression remained unchanged. Recombinant Ang-4 protein protected endothelial cells against serum starvation-induced apoptosis and increased cultured endothelial cell migration and tube formation. Ad2/HIF-1&agr;/VP16 stimulated endothelial cell proliferation and tube formation. Hypoxia- or Ad2/HIF-1&agr;/VP16-induced tube formation was significantly reduced by a Tie-2 inhibitor. These results suggest that HIF-1 mediates the angiogenic response to hypoxia by upregulating the expression of multiple angiogenic factors. Ang-4 can function similarly as Ang-1 and substitute for Ang-1 to participate in hypoxia-induced angiogenesis. Activation of the angiopoietin/Tie-2 system may play a role in the ability of HIF-1 to induce hypervascularity without excessive permeability.


Molecular Pharmacology | 2006

A Constitutively Active Hypoxia-Inducible Factor-1α/VP16 Hybrid Factor Activates Expression of the Human B-Type Natriuretic Peptide Gene

Yuxia Luo; Canwen Jiang; Adam J. Belanger; Geoffrey Y. Akita; Samuel C. Wadsworth; Richard J. Gregory; Karen A. Vincent

Hypoxia-inducible factor-1 (HIF-1) is a primary regulator of the physiological response to hypoxia. A recombinant adenovirus expressing a constitutively active hybrid form of the HIF-1α subunit (Ad2/HIF-1α/VP16) is being evaluated as a gene therapy for the treatment of peripheral vascular disease. Ad2/HIF-1α/VP16 up-regulates known HIF-1-responsive genes, including those involved in angiogenesis. Expression profile analysis revealed that the brain natriuretic peptide (BNP) gene was significantly up-regulated in response to HIF-1α/VP16 in human fetal cardiac cells. Real-time reverse transcription-polymerase chain reaction analyses confirmed transcriptional activation of the BNP gene by HIF-1α/VP16 in human but not rat cardiac cells. Because hypoxia itself did not increase human BNP gene expression in these analyses, the mechanism of the HIF-1α/VP16 effect was determined. Analyses of promoter deletion mutants suggested that the cis-acting sequence in the human BNP promoter mediating activation by HIF-1α/VP16 was a putative HIF-1 responsive element (HRE) located at -466. An SV40 basal promoter-luciferase plasmid containing a minimal BNP HRE was up-regulated by HIF-1α/VP16, whereas a similar construct carrying a mutation within the HIF-1 binding site was not. Mutation of an E-box motif within the BNP HRE reduced HIF-1α/VP16-mediated transcriptional activation by 50%. Gel-shift analyses showed that both the native HIF-1α and HIF-1α/VP16 are able to bind to a probe containing the HIF-1 binding site. These experiments demonstrate the existence of a functional HRE in the BNP promoter and further define the scope and mechanism of action of Ad2/HIF-1α/VP16.


Cell Transplantation | 2010

Factors affecting residence time of mesenchymal stromal cells (MSC) injected into the myocardium.

Jason Westrich; Peter C. Yaeger; Chufa He; Jeff Stewart; Raymond Chen; Gitta Seleznik; Shane Larson; Bruce M. Wentworth; Michael O'Callaghan; Sam Wadsworth; Geoffrey Y. Akita; Gyongyi Molnar

The therapeutic mechanism of mesenchymal stromal/stem cells (MSC) for the treatment of acute myocardial infarction is not well understood. Our goal was to get insights into this mechanism by analyzing the survival kinetics of allogeneic and syngeneic cell transplants under different tissue conditions. Two MSC cell banks, stably and equally expressing the luciferase reporter construct, were developed for these studies and injected directly to the myocardium of Lewis rat recipients under syngeneic or allogeneic transplantation conditions. Cell survival was monitored by real-time fashion for up to 2 weeks, using optical imaging device (IVIS, Xenogen Corp.). We found that both syngeneic and allogeneic grafts reduced significantly in size during the first week of transplantation, either in the normal or in the late infarcted heart (5 days after MI) and allotransplants became always smaller than syngeneic grafts during this period. Low dose of cyclosporine A treatment had a benefit on both allo- and syngeneic graft sizes, suggesting that multiple mechanisms play a role in early graft reduction. The MSC characteristic factors IL-6, IL-8, MCP-1, and VEGF were well above the control level in the heart tissue at 4 days after cell injection, suggesting that the peak therapeutic effect of MSC can be expected during the first week of the administration. Although allogeneic cells induced immunoglobulin production, their biological effects (cell survival, factor productions) are very similar to the syngeneic transplants and therefore they could deliver the same therapeutic effect as the syngeneic cells. Finally, freshly infarcted tissue (30 min) supported better the survival of MSC than late postischemic tissue (5 days) but only “off the shelf” allogeneic cell transplants fits with this treatment strategy.


Peptides | 2012

Cardiovascular effects of a PEGylated apelin.

Zhiqiang Jia; L. Hou; A. Leger; I. Wu; A.B. Kudej; J. Stefano; C. Jiang; C.Q. Pan; Geoffrey Y. Akita

Several studies have documented cardiovascular effects of apelin, including enhanced inotropy and vasodilation. However, these cardiovascular effects are short lived due to the predicted short circulating half-life of the apelin peptide. To address this limitation of apelin, we pursued N-terminal PEGylation of apelin and examined the cardiovascular effects of the PEGylated apelin. A 40kDa PEG conjugated apelin-36 (PEG-apelin-36) was successfully produced with N-terminal conjugation, high purity (>98%) and minimum reduction of APJ receptor binding affinity. Using an adenylate cyclase inhibition assay, comparable in vitro bioactivity was observed between the PEG-apelin-36 and unmodified apelin-36. In vivo evaluation of the PEG-apelin-36 was performed in normal rats and rats with myocardial infarction (MI). Cardiac function was assessed via echocardiography before, during a 20 min IV infusion and up to 100 min post peptide infusion. Similar increases in cardiac ejection fraction (EF) were observed during the infusion of PEG-apelin-36 and apelin-36 in normal rats. However, animals that received PEG-apelin-36 maintained significantly increased EF over the 100 min post infusion monitoring period compared to the animals that received unmodified apelin-36. Interestingly, EF increases observed with PEG-apelin-36 and apelin-36 were greater in the MI rats. PEG-apelin-36 had a prolonged circulating life compared to apelin-36 in rats. There were no changes in aortic blood pressure when PEG-apelin-36 or apelin-36 was administered. To our knowledge this is the first report of apelin PEGylation and documentation of its cardiovascular effects.


Journal of Gene Medicine | 2009

A hypoxic inducible factor-1α hybrid enhances collateral development and reduces vascular leakage in diabetic rats

Hidetoshi Kajiwara; Zhengyu Luo; Adam J. Belanger; Akihiro Urabe; Karen A. Vincent; Geoffrey Y. Akita; Seng H. Cheng; Seibu Mochizuki; Richard J. Gregory; Canwen Jiang

Diabetes mellitus is a common comorbidity of atherosclerosis. Hypoxia‐inducible factor‐1 (HIF‐1) is the master regulator of the angiogenic response to hypoxia.


Human Gene Therapy | 2001

Enhancement of Fas ligand-induced inhibition of neointimal formation in rabbit femoral and iliac arteries by coexpression of p35

Zhengyu Luo; Tracy Garron; Maria Palasis; Hsienwie Lu; Adam J. Belanger; Abraham Scaria; Karen A. Vincent; Taro Date; Geoffrey Y. Akita; Seng H. Cheng; James J. Barry; Richard J. Gregory; Canwen Jiang

Adenovirus-mediated gene transfer of Fas ligand (FasL) inhibits neointimal formation in balloon-injured rat carotid arteries. Vascular smooth muscle (VSM) cells coexpressing murine FasL and p35, a baculovirus gene that inhibits caspase activity, are not susceptible to FasL-mediated apoptosis in vitro but are capable of inducing apoptosis of VSM cells that do not express p35. We reasoned that coexpression of p35 in FasL-transduced VSM cells in vivo would promote their survival, enhance FasL-induced apoptosis of adjacent VSM cells, and thereby facilitate a greater inhibition of neointimal formation. In balloon-injured rabbit femoral arteries, either Ad2/FasL/p35 or Ad2/FasL was infused into the injured site and withdrawn 20 min later. Both vectors induced a dose-dependent reduction (p < 0.05) of the neointima-to-media ratio when assessed 14 days later. However, Ad2/FasL/p35 exhibited a significantly greater inhibition of neointimal formation than Ad2/FasL. In a more clinically relevant model of restenosis, rabbit iliac arteries were injured with an angioplasty catheter under fluoroscopic guidance. Adenoviral vectors were delivered locally to the injured site over a period of 2 min, using a porous infusion balloon catheter. Twenty-eight days after gene transfer angiographic and histologic assessments indicated a significant (p < 0.05) inhibition of iliac artery lumen stenosis and neointimal formation by Ad2/FasL/p35 (5 x 10(11) particles per artery). The extent of inhibition was comparable to that achieved with Ad2/TK, an adenoviral vector encoding thymidine kinase (5 x 10(11) particles per artery) and coadministration of ganciclovir for 7 days. These data suggest that coexpression of p35 in FasL-transduced VSM cells is more potent at inhibiting neointimal formation and as such represents an improved gene therapy approach for restenosis.


Circulation | 2005

Adenovirus-Mediated Expression of β-Adrenergic Receptor Kinase C-Terminus Reduces Intimal Hyperplasia and Luminal Stenosis of Arteriovenous Polytetrafluoroethylene Grafts in Pigs

Zhengyu Luo; Geoffrey Y. Akita; Taro Date; Christopher M. Treleaven; Karen A. Vincent; Denise Woodcock; Seng H. Cheng; Richard J. Gregory; Canwen Jiang

Background—Hemodialysis vascular access dysfunction is the single most important cause of morbidity in kidney hemodialysis patients. Failure of an arteriovenous polytetrafluoroethylene (PTFE) graft, the most common form of hemodialysis access, is primarily due to intimal hyperplasia and thrombosis at the venous anastomosis. Methods and Results—This study was aimed at evaluating the efficacy and safety of an adenoviral vector (Ad2/&bgr;ARKct) encoding the carboxyl terminus of &bgr;-adrenergic receptor kinase (&bgr;ARKct) in a pig model of arteriovenous PTFE graft failure. Transduction of the external jugular vein with Ad2/&bgr;ARKct (5E9, 5E10, or 5E11 particles per vein) did not result in systemic toxicity, as measured by clinical and pathological assessments. Ad2/&bgr;ARKct significantly reduced neointimal hyperplasia in the graft/vein anastomosis. It also improved the graft patency rate and angiographic score, as measured histologically and angiographically, compared with vehicle or empty viral vector controls. Conclusions—Our results suggest that local administration of adenoviral vectors encoding &bgr;ARKct into the jugular vein represents a viable strategy to treat AV graft hemodialysis vascular access failure.


Molecular Pharmacology | 2006

Potential therapeutic gene for the treatment of ischemic disease: Ad2/HIF-1α/VP16 enhances BNP gene expression via an HIF-1 responsive element.

Yuxia Luo; Canwen Jiang; Adam J. Belanger; Geoffrey Y. Akita; Samuel C. Wadsworth; Karen A. Vincent; Michael E. Wilhide; W. Keith Jones

In this issue of Molecular Pharmacology, Luo et al. (p. 1953) present a study employing a HIF-1α/VP16 chimera to investigate the mechanism by which this constitutively active transcription factor activates expression of brain natriuretic peptide (BNP). The results define a functional hypoxia responsive element (HRE) in the promoter of the human BNP gene and demonstrate that this HRE is necessary for HIF-1α/VP16-induced gene expression in human cardiomyocytes grown under normoxic conditions. Luo et al. also show that a consensus E-box DNA binding sequence is necessary for appropriate BNP regulation. Because HIF-1 is known to elicit protective and beneficial gene expression programs in many scenarios and because BNP is known to be cardioprotective, this study provides support for the therapeutic use of the chimeric HIF-1α/VP16 protein in coronary heart disease. However, because HIF-1α is a key regulatory molecule that acts upon a large number of downstream gene networks, there remains a need for further investigation. Particularly useful would be comprehensive gene expression profiling coupled with functional analysis of HIF-1α/VP16-regulated genes. The results of such studies will elucidate the mechanism of beneficial effects and address concerns regarding potential adverse effects of activating specific HIF-1α/VP16-dependent gene programs.


Journal of Vascular and Interventional Radiology | 2000

Angiogenesis Is Induced in a Rabbit Model of Hindlimb Ischemia by Naked DNA Encoding a HIF-la/VP16 Hybrid Transcription Factor

Karen A. Vincent; Kou-Gi Shyu; Yuxia Luo; Meredith Magner; Rene A. Tio; Canwen Jiang; Mark A. Goldberg; Geoffrey Y. Akita; Jeffrey M. Isner; Richard J. Gregory

BACKGROUND Hypoxia-inducible factor-1 (HIF-1) is a heterodimeric transcription factor that regulates expression of genes involved in O(2) homeostasis, including vascular endothelial growth factor (VEGF), a potent stimulator of angiogenesis. We sought to exploit this native adaptive response to hypoxia as a treatment for chronic ischemia. METHODS AND RESULTS A hybrid protein consisting of DNA-binding and dimerization domains from the HIF-1alpha subunit and the transactivation domain from herpes simplex virus VP16 protein was constructed to create a strong, constitutive transcriptional activator. After transfection into HeLa, C6, and Hep3B cells, this chimeric transcription factor was shown to activate expression of the endogenous VEGF gene, as well as several other HIF-1 target genes in vitro. The bioactivity of HIF-1alpha/VP16 hybrid gene transfer in vivo was examined in a rabbit model of hindlimb ischemia. Administration of HIF-1alpha/VP16 was associated with significant improvements in calf blood pressure ratio, angiographic score, resting and maximal regional blood flow, and capillary density (all P:<0.01). CONCLUSIONS The HIF-1alpha/VP16 hybrid transcription factor is able to promote significant improvement in perfusion of an ischemic limb. These results confirm the feasibility of a novel approach for therapeutic angiogenesis in which neovascularization may be achieved indirectly by use of a transcriptional regulatory strategy.

Collaboration


Dive into the Geoffrey Y. Akita's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amelia B. Kudej

University of Medicine and Dentistry of New Jersey

View shared research outputs
Researchain Logo
Decentralizing Knowledge