Network


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

Hotspot


Dive into the research topics where Larry L. Shears is active.

Publication


Featured researches published by Larry L. Shears.


American Journal of Physiology-heart and Circulatory Physiology | 1999

Impaired wound healing and angiogenesis in eNOS-deficient mice

Paul C. Lee; A.Neil Salyapongse; Gwynne A. Bragdon; Larry L. Shears; Simon C. Watkins; Howard D. Edington; Timothy R. Billiar

A role for nitric oxide (NO) in wound healing has been proposed; however, the absolute requirement of NO for wound healing in vivo and the contribution of endothelial NO synthase (eNOS) have not been determined. Experiments were carried out using eNOS gene knockout (KO) mice to determine the requirement for eNOS on wound closure and wound strength. Excisional wound closure was significantly delayed in the eNOS KO mice (29.4 +/- 2.2 days) compared with wild-type (WT) controls (20.2 +/- 0.4 days). At 10 days, incisional wound tensile strength demonstrated a 38% reduction in the eNOS KO mice. Because effective wound repair requires growth factor-stimulated angiogenesis, in vitro and in vivo angiogenesis assays were performed in the mice to assess the effects of eNOS deficiency on angiogenesis. Endothelial cell sprouting assays confirmed in vitro that eNOS is required for proper endothelial cell migration, proliferation, and differentiation. Aortic segments harvested from eNOS KO mice cultured with Matrigel demonstrated a significant reduction in endothelial cell sprouting and [(3)H]thymidine incorporation compared with WT mice at 5 days. Capillary ingrowth into subcutaneously implanted Matrigel plugs was significantly reduced in eNOS KO mice (2.67 +/- 0.33 vessels/plug) compared with WT mice (10.17 +/- 0.79 vessels/plug). These results clearly show that eNOS plays a significant role in facilitating wound repair and growth factor-stimulated angiogenesis.A role for nitric oxide (NO) in wound healing has been proposed; however, the absolute requirement of NO for wound healing in vivo and the contribution of endothelial NO synthase (eNOS) have not been determined. Experiments were carried out using eNOS gene knockout (KO) mice to determine the requirement for eNOS on wound closure and wound strength. Excisional wound closure was significantly delayed in the eNOS KO mice (29.4 ± 2.2 days) compared with wild-type (WT) controls (20.2 ± 0.4 days). At 10 days, incisional wound tensile strength demonstrated a 38% reduction in the eNOS KO mice. Because effective wound repair requires growth factor-stimulated angiogenesis, in vitro and in vivo angiogenesis assays were performed in the mice to assess the effects of eNOS deficiency on angiogenesis. Endothelial cell sprouting assays confirmed in vitro that eNOS is required for proper endothelial cell migration, proliferation, and differentiation. Aortic segments harvested from eNOS KO mice cultured with Matrigel demonstrated a significant reduction in endothelial cell sprouting and [3H]thymidine incorporation compared with WT mice at 5 days. Capillary ingrowth into subcutaneously implanted Matrigel plugs was significantly reduced in eNOS KO mice (2.67 ± 0.33 vessels/plug) compared with WT mice (10.17 ± 0.79 vessels/plug). These results clearly show that eNOS plays a significant role in facilitating wound repair and growth factor-stimulated angiogenesis.


Journal of Clinical Investigation | 1997

Inducible nitric oxide synthase suppresses the development of allograft arteriosclerosis.

Larry L. Shears; Nobuyoshi Kawaharada; Edith Tzeng; Timothy R. Billiar; Simon C. Watkins; Imre Kovesdi; Alena Lizonova; Si M. Pham

In cardiac transplantation, chronic rejection takes the form of an occlusive vasculopathy. The mechanism underlying this disorder remains unclear. The purpose of this study was to investigate the role nitric oxide (NO) may play in the development of allograft arteriosclerosis. Rat aortic allografts from ACI donors to Wistar Furth recipients with a strong genetic disparity in both major and minor histocompatibility antigens were used for transplantation. Allografts collected at 28 d were found to have significant increases in both inducible NO synthase (iNOS) mRNA and protein as well as in intimal thickness when compared with isografts. Inhibiting NO production with an iNOS inhibitor increased the intimal thickening by 57.2%, indicating that NO suppresses the development of allograft arteriosclerosis. Next, we evaluated the effect of cyclosporine (CsA) on iNOS expression and allograft arteriosclerosis. CsA (10 mg/kg/d) suppressed the expression of iNOS in response to balloon-induced aortic injury. Similarly, CsA inhibited iNOS expression in the aortic allografts, associated with a 65% increase in intimal thickening. Finally, we investigated the effect of adenoviral-mediated iNOS gene transfer on allograft arteriosclerosis. Transduction with iNOS using an adenoviral vector suppressed completely the development of allograft arteriosclerosis in both untreated recipients and recipients treated with CsA. These results suggest that the early immune-mediated upregulation in iNOS expression partially protects aortic allografts from the development of allograft arteriosclerosis, and that iNOS gene transfer strategies may prove useful in preventing the development of this otherwise untreatable disease process.


Journal of The American College of Surgeons | 1998

Efficient inhibition of intimal hyperplasia by adenovirus-mediated inducible nitric oxide synthase gene transfer to rats and pigs in vivo

Larry L. Shears; Melina R. Kibbe; Alan Murdock; Timothy R. Billiar; Alena Lizonova; Imre Kovesdi; Simon C. Watkins; Edith Tzeng

BACKGROUND Inadequate nitric oxide (NO) availability may underlie vascular smooth muscle overgrowth that contributes to vascular occlusive diseases including atherosclerosis and restenosis. NO possesses a number of properties that should inhibit this hyperplastic healing response, such as promoting reendothelialization, preventing platelet and leukocyte adherence, and inhibiting cellular proliferation. STUDY DESIGN We proposed that shortterm but sustained increases in NO synthesis achieved with inducible NO synthase (iNOS) gene transfer at sites of vascular injury would prevent intimal hyperplasia. We constructed an adenoviral vector, AdiNOS, carrying the human iNOS cDNA and used it to express iNOS at sites of arterial injury in vivo. RESULTS AdiNOS-treated cultured vascular smooth muscle cells produced up to 100-fold more NO than control cells. In vivo iNOS gene transfer, using low concentrations of AdiNOS (2 x 10(6) plaque forming units [PFU]/rat) to injured rat carotid arteries, resulted in a near complete (>95%) reduction in neointima formation even when followed longterm out to 6 weeks post-injury. This protective effect was reversed by the continuous administration of an iNOS selective inhibitor L-N6-(1-iminoethyl)-lysine. However, iNOS gene transfer did not lead to regression of preestablished neointimal lesions. In an animal model more relevant to human vascular healing, iNOS gene transfer (5 x 10(8) PFU/pig) to injured porcine iliac arteries in vivo was also efficacious, reducing intimal hyperplasia by 51.8%. CONCLUSIONS These results indicate that shortterm overexpression of the iNOS gene initiated at the time of vascular injury is an effective method of locally increasing NO levels to prevent intimal hyperplasia.


Transplantation | 2000

Endothelial nitric oxide synthase protects aortic allografts from the development of transplant arteriosclerosis

Paul C. Lee; Zhi Liang Wang; Shiguang Qian; Simon C. Watkins; Alena Lizonova; Imre Kovesdi; Edith Tzeng; Richard L. Simmons; Timothy R. Billiar; Larry L. Shears

BACKGROUND Inducible nitric oxide synthase (iNOS) is up-regulated in rejecting allografts and is protective against allograft arteriosclerosis; it suppresses neointimal smooth muscle cell accumulation and inhibits adhesion of platelets and leukocytes to the endothelium. However, the functional importance of endothelial NOS (eNOS) in the rejecting allografts remains unclear. METHODS We examined the effects of selective eNOS deficiency in aortic allografts in a murine chronic rejection model using grafts from eNOS knockout (KO) mice (C57BL/6 background; H2b) and normal C3H (H2K) as recipients. Grafts from wild-type C57BL/6 mice served as controls. Grafts from iNOS KO mice served as a second group of controls where the contribution from iNOS was eliminated but eNOS was preserved. Aortic grafts were harvested and analyzed at days 10-14, 18-22, and 26-30 after transplantation. RESULTS Endothelial NOS-deficient grafts showed significantly increased intima/media ratios at days 26-30 compared to controls. Immunostaining demonstrated that in eNOS KO grafts, eNOS was not detectable whereas iNOS was expressed prominently in infiltrating recipient mononuclear cells. In control grafts, eNOS expression was preserved in the endothelium even by day 30, and associated with a decrease in intimal thickening. We further demonstrated that early overexpression of iNOS by ex vivo gene transfer completely prevented the development of arteriosclerosis associated with eNOS deficiency. CONCLUSIONS We found that eNOS plays a protective role in allografts, and that in eNOS-deficient allografts, early overexpression of iNOS is capable of preventing the development of allograft arteriosclerosis. In allografts with dysfunctional vascular endothelium and impaired eNOS activity as a result of ischemia or native arteriosclerotic disease, iNOS gene therapy may serve to improve their long-term survival and function.


Surgery | 1996

Vascular inducible nitric oxide synthase gene therapy: Requirement for guanosine triphosphate cyclohydrolase I

Edith Tzeng; Toshie Yoneyama; Kazuyuki Hatakeyama; Larry L. Shears; Timothy R. Billiar

BACKGROUND Human inducible nitric oxide synthase (iNOS) gene transfer inhibits myointimal hyperplasia in vitro. However, unstimulated vascular smooth muscle cells (SMC) do not synthesize tetrahydrobiopterin (BH4), an essential cofactor for iNOS, which may be an obstacle to successful vascular iNOS gene therapy. We investigated the capacity of gene transfer of guanosine triphosphate (GTP) cyclohydrolase I (GTPCH), the rate-limiting enzyme for BH4 biosynthesis, to supply cofactor for iNOS activity. METHODS A human GTPCH expression plasmid (pCIS-GTPCH) was transfected into rat aortic SMC (RAOSMC) and BH4-deficient NIH3T3 cells engineered to stably express human iNOS (3T3-iNOS). GTPCH activity and intracellular biopterins were assessed as a measure of successful transfection, and the capacity of GTPCH to reconstitute iNOS activity was used to determine whether BH4 was made available to the iNOS protein. RESULTS The pCIS-GTPCH-transfected 3T3 cells had demonstrable GTPCH activity as compared with control cells (169.3 +/- 6.6 pmol/hr/mg versus 0, p < 0.001). Intracellular biopterin levels were also increased in transfected 3T3 and SMC (60.6 +/- 2.6 and 101.7 +/- 28.3 pmol/mg, respectively, versus less than 4 in control cells). GTPCH reconstituted near-maximal iNOS activity in 3T3-iNOS cells despite a gene transfer efficiency of less than 1%. GTPCH and iNOS enzymes did not have to coexist in the same cell for the synthesized BH4 to support iNOS activity. CONCLUSION GTPCH gene transfer reconstitutes iNOS activity in BH4-deficient cells despite poor transfer efficiency. GTPCH can deliver a cofactor to targeted cells even if it is synthesized in neighboring cells, and may be a means to concurrently deliver BH4 with iNOS in vivo.


Journal of Heart and Lung Transplantation | 1999

Mixed hematopoietic chimerism prevents allograft vasculopathy

Nobuyoshi Kawaharada; Larry L. Shears; Sen Li; Si M. Pham

BACKGROUND Mixed hematopoietic chimerism has been shown to induce long-term acceptance of transplant organs. We determined whether mixed chimerism prevented allograft vasculopathy, using the rat aortic allograft model. METHODS Mixed chimeras were prepared by reconstituting lethally irradiated (1100 cGy) WF rats with a mixture of T-cell depleted (TCD) syngeneic (WF) plus TCD allogeneic (ACI) bone marrow. Donor-specific (ACI) or third-party (F344) aortic grafts were transplanted into mixed chimeric animals 1 to 2 months after bone marrow reconstitution. No immunosuppressive drugs were administered. At 30 days postoperatively, aortic allografts were harvested for histology and measurement of cytokine mRNA by semiquantitative RT-PCR. Some aortic grafts were harvested at 90 and 180 days after transplantation for histological analysis. The degree of intimal hyperplasia and cytokine gene expression were compared among 4 groups: I (syngeneic; ACI donors to ACI recipients), II (allografts; ACI to WF), III (donor specific; ACI donor to chimeras) and IV (third-party; F344 to chimeras). RESULTS There was no difference in the degree of intimal hyperplasia (IH) between groups I and III. Groups II and IV had significantly more IH than group I. Compared to group I, levels of mRNA for IFN-y, IL-2, IL-10 and iNOS in groups II and IV were higher, while there was no difference in mRNA levels between group I and III. CONCLUSIONS These data suggest that mixed chimerism prevents allograft vasculopathy. Mixed chimerism holds great promise in clinical transplantation as a means to prevent allograft vasculopathy.


Clinical and Experimental Pharmacology and Physiology | 1999

ROLE OF INDUCIBLE NITRIC OXIDE SYNTHASE IN TRANSPLANT ARTERIOSCLEROSIS

Paul C. Lee; Larry L. Shears; Timothy R. Billiar

1. Transplant arteriosclerosis is a major obstacle to long‐term allograft survival. Nitric oxide (NO) has been implicated as a mediator in the development of this disease.


Journal of Heart and Lung Transplantation | 2003

Hearts transfected with inducible nitric oxide synthase have accelerated restoration of cardiac function following ischemia/reperfusion associated with cardiac transplantation

Larry L. Shears; Shinichi Kanno; Timothy R. Billiar; R. Mahidara; Kenneth R. McCurry; Robert L. Kormos

Abstract Purpose: Our laboratory has demonstrated the novel effect of iNOS gene therapy in preventing the development of cardiac allograft vasculopathy. The effect of this therapeutic strategy on graft function needs to be evaluated prior to initiating clinical trials. This study documents the effect that iNOS transfection has on graft function following a period of ischemia/reperfusion similar to what would be incurred during cardiac transplantation. Procedure: Isografts were transfected via continuous antegrade coronary perfusion with either AdiNOS or the reporter gene AdEGFP (5X 10 8 pfu)prior to heterotopic transplantation. Grafts were harvested 4 days later and placed on a Langendorf apparatus. Grafts were then subjected to 20 minutes of warm ischemia and then reperfused. Coronary blood flow (CBF), contractility (dP/dT), diastolic relaxation (-dP/dT), and left ventricular pressures (LVP) were recorded at 20 and 120 minute intervals after reperfusion. Results: As shown in the table below, grafts transfected with AdiNOS had a significant restoration in contractility, diastolic relaxation and coronary blood flow at 120 minutes when compared to their respective AdEGFP controls. ∗ . Pre EGFP Post EGFP 20 minutes Post EGFP 120 minutes Pre iNOS Post iNOS 20 minutes Post iNOS 120 minutes CF 12.03 ± 0.61 6.15 ± 0.44 4.28 ± 0.35 12.180 ± 0.74 5.75 ± 0.21 9.08 ± 0.61 ∗ dP/dT 2503 ± 822 1202 ± 129 1156 ± 470 2075 ± 511 960 ± 46.5 1826 ± 479 ∗ −dP/dT −1959 ± 591 −792 ± 102 −691 ± 201 −1475 ± 432 −630 ± 107 −1320.2 ± 624 ∗ LVP 107.7 ± 11.4 41.5 ± 5.3 42.3 ± 7.1 87.0 ± 10.7 34.8 ± 3.0 69.5 ± 10.1 ∗ ∗ p Conclusions: AdiNOS gene therapy accelerates the restoration of baseline cardiac function following ischemia/reperfusion. The mechanism by which nitric oxide ameliorates the toxic effect of ischemia/reperfusion needs to be further studied. These data suggest further evaluation of this therapeutic strategy to prevent allograft vasculopathy is warranted and support the initiation of clinical trials.


Journal of Virology | 1997

INCREASED IN VITRO AND IN VIVO GENE TRANSFER BY ADENOVIRUS VECTORS CONTAINING CHIMERIC FIBER PROTEINS

Thomas J. Wickham; Edith Tzeng; Larry L. Shears; Peter W. Roelvink; Gai M. Lee; Douglas E. Brough; Alena Lizonova; Imre Kovesdi


Proceedings of the National Academy of Sciences of the United States of America | 2004

Nitric oxide facilitates cardiomyogenesis in mouse embryonic stem cells

Shinichi Kanno; Peter K. Kim; Karim Sallam; Jing Lei; Timothy R. Billiar; Larry L. Shears

Collaboration


Dive into the Larry L. Shears's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Edith Tzeng

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul C. Lee

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John M. Kane

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Melina R. Kibbe

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge