Tumstatin

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 798 Experts worldwide ranked by ideXlab platform

Raghu Kalluri - One of the best experts on this subject based on the ideXlab platform.

  • Penetration of Endothelial Cell Coated Multicellular Tumor Spheroids by Iron Oxide Nanoparticles
    Theranostics, 2012
    Co-Authors: Nathan Kohler, Raghu Kalluri, Aruna Sigdel, Jeffrey R. Morgan, Shouheng Sun
    Abstract:

    Iron oxide nanoparticles are a useful diagnostic contrast agent and have great potential for therapeutic applications. Multiple emerging diagnostic and therapeutic applications and the numerous versatile parameters of the nanoparticle platform require a robust biological model for characterization and assessment. Here we investigate the use of iron oxide nanoparticles that target tumor vasculature, via the Tumstatin peptide, in a novel three-dimensional tissue culture model. The developed tissue culture model more closely mimics the in vivo environment with a leaky endothelium coating around a glioma tumor mass. Tumstatin-iron oxide nanoparticles showed penetration and selective targeting to endothelial cell coating on the tumor in the three-dimensional model, and had approximately 2 times greater uptake in vitro and 2.7 times tumor neo-vascularization inhibition. Tumstatin provides targeting and therapeutic capabilities to the iron oxide nanoparticle diagnostic contrast agent platform. And the novel endothelial cell-coated tumor model provides an in vitro microtissue environment to evaluate nanoparticles without moving into costly and time-consuming animal models.

  • Penetration of Endothelial Cell Coated Multicellular Tumor Spheroids by Iron Oxide Nanoparticles
    2012
    Co-Authors: Nathan Kohler, Raghu Kalluri, Aruna Sigdel, Jeffrey R. Morgan, Shouheng Sun
    Abstract:

    licenses/by-nc-nd/3.0/). Reproduction is permitted for personal, noncommercial use, provided that the article is in whole, unmodified, and properly cited. Received: 2011.09.28; Accepted: 2011.10.24; Published: 2012.01.01 Iron oxide nanoparticles are a useful diagnostic contrast agent and have great potential for therapeutic applications. Multiple emerging diagnostic and therapeutic applications and the numerous versatile parameters of the nanoparticle platform require a robust biological model for characterization and assessment. Here we investigate the use of iron oxide nanoparticles that target tumor vasculature, via the Tumstatin peptide, in a novel three-dimensional tissue culture model. The developed tissue culture model more closely mimics the in vivo environment with a leaky endothelium coating around a glioma tumor mass. Tumstatin-iron oxide nanoparticles showed penetration and selective targeting to endothelial cell coating on the tumor in the three-dimensional model, and had approximately 2 times greater uptake in vitro and 2.7 times tumor neo-vascularization inhibition. Tumstatin provides targeting and therapeutic capabilities to the iron oxide nanoparticle diagnostic contrast agent platform. And th

  • identification of amino acids essential for the antiangiogenic activity of Tumstatin and its use in combination antitumor activity
    Proceedings of the National Academy of Sciences of the United States of America, 2008
    Co-Authors: Hans Petter Eikesdal, Yohei Maeshima, Hikaru Sugimoto, Gabriel Birrane, Vesselina G Cooke, Mark W Kieran, Raghu Kalluri
    Abstract:

    Tumstatin is an angiogenesis inhibitor that binds to αvβ3 integrin and suppresses tumor growth. Previous deletion mutagenesis studies identified a 25-aa fragment of Tumstatin (Tumstatin peptide) with in vitro antiangiogenic activity. Here, we demonstrate that systemic administration of this Tumstatin peptide inhibits tumor growth and angiogenesis. Site-directed mutagenesis identified amino acids L, V, and D as essential for the antiangiogenic activity of Tumstatin. The Tumstatin peptide binds to αvβ3 integrin on proliferating endothelial cells and localizes to select tumor endothelium in vivo. Using 3D molecular modeling, we identify a putative interaction interface for Tumstatin peptide on αvβ3 integrin. The antitumor activity of the Tumstatin peptide, in combination with bevacizumab (anti-VEGF antibody), displays significant improvement in efficacy against human renal cell carcinoma xenografts when compared with the single-agent use. Collectively, our results demonstrate that Tumstatin peptide binds specifically to the tumor endothelium, and its antiangiogenic action is mediated by αvβ3 integrin, and, in combination with an anti-VEGF antibody it exhibits enhanced tumor suppression of renal cell carcinoma.

  • Tumstatin, the NC1 domain of α3 chain of type IV collagen, is an endogenous inhibitor of pathological angiogenesis and suppresses tumor growth
    Biochemical and biophysical research communications, 2005
    Co-Authors: Yuki Hamano, Raghu Kalluri
    Abstract:

    Angiogenesis, the formation of new blood vessels, is required for physiological development of vertebrates and repair of damaged tissue, but in the pathological setting contributes to progression of cancer. During tumor growth, angiogenesis is supported by up-regulation of angiogenic stimulators (pro-angiogenic) and down-regulation of angiogenic inhibitors (anti-angiogenic). The switch to the angiogenic phenotype (angiogenic switch) allows the tumors to grow and facilitate metastasis. The bioactive NC1 domain of type IV collagen α3 chain, called Tumstatin, imparts anti-tumor activity by inducing apoptosis of proliferating endothelial cells. Tumstatin binds to αVβ3 integrin via a mechanism independent of the RGD-sequence recognition and inhibits cap-dependent protein synthesis in the proliferating endothelial cells. The physiological level of Tumstatin is controlled by matrix metalloproteinase-9, which most effectively cleaves it from the basement membrane and its physiological concentration in the circulation keeps pathological angiogenesis and tumor growth in check. These findings suggest that Tumstatin functions as an endogenous inhibitor of pathological angiogenesis and functions as a novel suppressor of proliferating endothelial cells and growth of tumors.

  • Human Tumstatin and human endostatin exhibit distinct antiangiogenic activities mediated by αvβ3 and α5β1 integrins
    Proceedings of the National Academy of Sciences of the United States of America, 2003
    Co-Authors: Akulapalli Sudhakar, Julie C Lively, Changqing Yang, Hikaru Sugimoto, Michael Zeisberg, Raghu Kalluri
    Abstract:

    Tumstatin and endostatin are two inhibitors of angiogenesis derived from precursor human collagen molecules known as α3 chain of type IV collagen and α1 chain of type XVIII collagen, respectively. Although both these inhibitors are noncollagenous (NC1) domain fragments of collagens, they only share a 14% amino acid homology. In the present study we evaluated the functional receptors, mechanism of action, and intracellular signaling induced by these two collagen-derived inhibitors. Human Tumstatin prevents angiogenesis via inhibition of endothelial cell proliferation and promotion of apoptosis with no effect on migration, whereas human endostatin prevents endothelial cell migration with no effect on proliferation. We demonstrate that human Tumstatin binds to αvβ3 integrin in a vitronectin/fibronectin/RGD cyclic peptide independent manner, whereas human endostatin competes with fibronectin/RGD cyclic peptide to bind α5β1 integrin. The activity of human Tumstatin is mediated by αvβ3 integrin, whereas the activity of human endostatin is mediated by α5β1 integrin. Additionally, although human Tumstatin binding to αvβ3 integrin leads to the inhibition of Cap-dependent translation (protein synthesis) mediated by focal adhesion kinase/phosphatidylinositol 3-kinase/Akt/mTOR/4E-BP1 pathway, human endostatin binding to α5β1 integrin leads to the inhibition of focal adhesion kinase/c-Raf/MEK1/2/p38/ERK1 mitogen-activated protein kinase pathway, with no effect on phosphatidylinositol 3-kinase/Akt/mTOR/4E-BP1 and Cap-dependent translation. Collectively, such distinct properties of human Tumstatin and human endostatin provide the first insight into their diverse antiangiogenic actions and argue for combining them for targeting tumor angiogenesis.

Janette K. Burgess - One of the best experts on this subject based on the ideXlab platform.

  • Tumstatin regulates the angiogenic and inflammatory potential of airway smooth muscle extracellular matrix
    Journal of cellular and molecular medicine, 2017
    Co-Authors: Louise M. Harkness, Markus Weckmann, Matthias Kopp, Tim Becker, Anthony W. Ashton, Janette K. Burgess
    Abstract:

    The extracellular matrix (ECM) creates the microenvironment of the tissue; an altered ECM in the asthmatic airway may be central in airway inflammation and remodelling. Tumstatin is a collagen IV-derived matrikine reduced in the asthmatic airway wall that reverses airway inflammation and remodelling in small and large animal models of asthma. This study hypothesized that the mechanisms underlying the broad asthma-resolving effects of Tumstatin were due to autocrine remodelling of the ECM. Neutrophils and endothelial cells were seeded on decellularized ECM of non-asthmatic (NA) or asthmatic (A) airway smooth muscle (ASM) cells previously exposed to Tumstatin in the presence or absence of a broad matrix metalloproteinase inhibitor, Marimastat. Gene expression in NA and A ASM induced by Tumstatin was assessed using RT-PCR arrays. The presence of Tumstatin during ECM deposition affected neutrophil and endothelial cell properties on both NA and A ASM-derived matrices and this was only partly due to MMP activity. Gene expression patterns in response to Tumstatin in NA and A ASM cells were different. Tumstatin may foster an anti-inflammatory and anti-angiogenic microenvironment by modifying ASM-derived ECM. Further work is required to examine whether restoring Tumstatin levels in the asthmatic airway represents a potential novel therapeutic approach.

  • The Effects of Tumstatin on Vascularity, Airway Inflammation and Lung Function in an Experimental Sheep Model of Chronic Asthma
    Scientific reports, 2016
    Co-Authors: Joanne Van Der Velden, Louise M. Harkness, Donna Barker, Garry Barcham, Cathryn L Ugalde, Emmanuel Koumoundouros, Heidi Bao, Louise Organ, Ana Tokanovic, Janette K. Burgess
    Abstract:

    Tumstatin, a protein fragment of the alpha-3 chain of Collagen IV, is known to be significantly reduced in the airways of asthmatics. Further, there is evidence that suggests a link between the relatively low level of Tumstatin and the induction of angiogenesis and inflammation in allergic airway disease. Here, we show that the intra-segmental administration of Tumstatin can impede the development of vascular remodelling and allergic inflammatory responses that are induced in a segmental challenge model of experimental asthma in sheep. In particular, the administration of Tumstatin to lung segments chronically exposed to house dust mite (HDM) resulted in a significant reduction of airway small blood vessels in the diameter range 10+–20 μm compared to controls. In Tumstatin treated lung segments after HDM challenge, the number of eosinophils was significantly reduced in parenchymal and airway wall tissues, as well as in the bronchoalveolar lavage fluid. The expression of VEGF in airway smooth muscle was also significantly reduced in Tumstatin-treated segments compared to control saline-treated segments. Allergic lung function responses were not attenuated by Tumstatin administration in this model. The data are consistent with the concept that Tumstatin can act to suppress vascular remodelling and inflammation in allergic airway disease.

  • LF-15 & T7, synthetic peptides derived from Tumstatin, attenuate aspects of airway remodelling in a murine model of chronic OVA-induced allergic airway disease.
    PloS one, 2014
    Co-Authors: Karryn T Grafton, Janette K. Burgess, Judith L Black, Lyn M. Moir, Nicole G. Hansbro, Philip M. Hansbro, Brian G Oliver
    Abstract:

    Background Tumstatin is a segment of the collagen-IV protein that is markedly reduced in the airways of asthmatics. Tumstatin can play an important role in the development of airway remodelling associated with asthma due to its anti-angiogenic properties. This study assessed the anti-angiogenic properties of smaller peptides derived from Tumstatin, which contain the interface Tumstatin uses to interact with the αVβ3 integrin. Methods Primary human lung endothelial cells were exposed to the LF-15, T3 and T7 Tumstatin-derived peptides and assessed for cell viability and tube formation in vitro. The impact of the anti-angiogenic properties on airways hyperresponsiveness (AHR) was then examined using a murine model of chronic OVA-induced allergic airways disease. Results The LF-15 and T7 peptides significantly reduced endothelial cell viability and attenuated tube formation in vitro. Mice exposed to OVA+ LF-15 or OVA+T7 also had reduced total lung vascularity and AHR was attenuated compared to mice exposed to OVA alone. T3 peptides reduced cell viability but had no effect on any other parameters. Conclusion The LF-15 and T7 peptides may be appropriate candidates for use as novel pharmacotherapies due to their small size and anti-angiogenic properties observed in vitro and in vivo.

  • lf 15 t7 synthetic peptides derived from Tumstatin attenuate aspects of airway remodelling in a murine model of chronic ova induced allergic airway disease
    PLOS ONE, 2014
    Co-Authors: Karryn T Grafton, Janette K. Burgess, Judith L Black, Lyn M. Moir, Nicole G. Hansbro, Philip M. Hansbro, Brian G Oliver
    Abstract:

    Background Tumstatin is a segment of the collagen-IV protein that is markedly reduced in the airways of asthmatics. Tumstatin can play an important role in the development of airway remodelling associated with asthma due to its anti-angiogenic properties. This study assessed the anti-angiogenic properties of smaller peptides derived from Tumstatin, which contain the interface Tumstatin uses to interact with the αVβ3 integrin. Methods Primary human lung endothelial cells were exposed to the LF-15, T3 and T7 Tumstatin-derived peptides and assessed for cell viability and tube formation in vitro. The impact of the anti-angiogenic properties on airways hyperresponsiveness (AHR) was then examined using a murine model of chronic OVA-induced allergic airways disease. Results The LF-15 and T7 peptides significantly reduced endothelial cell viability and attenuated tube formation in vitro. Mice exposed to OVA+ LF-15 or OVA+T7 also had reduced total lung vascularity and AHR was attenuated compared to mice exposed to OVA alone. T3 peptides reduced cell viability but had no effect on any other parameters. Conclusion The LF-15 and T7 peptides may be appropriate candidates for use as novel pharmacotherapies due to their small size and anti-angiogenic properties observed in vitro and in vivo.

  • LF-15 & T7, Synthetic Peptides Derived from Tumstatin, Attenuate Aspects of Airway Remodelling in a Murine Model of Chronic OVA-Induced Allergic Airway Disease
    2014
    Co-Authors: Karryn T Grafton, Janette K. Burgess, Judith L Black, Lyn M. Moir, Nicole G. Hansbro, Philip M. Hansbro, Brian G Oliver
    Abstract:

    Background: Tumstatin is a segment of the collagen-IV protein that is markedly reduced in the airways of asthmatics. Tumstatin can play an important role in the development of airway remodelling associated with asthma due to its anti-angiogenic properties. This study assessed the anti-angiogenic properties of smaller peptides derived from Tumstatin, which contain the interface Tumstatin uses to interact with the aVb3 integrin. Methods: Primary human lung endothelial cells were exposed to the LF-15, T3 and T7 Tumstatin-derived peptides and assessed for cell viability and tube formation in vitro. The impact of the anti-angiogenic properties on airways hyperresponsiveness (AHR) was then examined using a murine model of chronic OVA-induced allergic airways disease. Results: The LF-15 and T7 peptides significantly reduced endothelial cell viability and attenuated tube formation in vitro. Mice exposed to OVA+ LF-15 or OVA+T7 also had reduced total lung vascularity and AHR was attenuated compared to mice exposed to OVA alone. T3 peptides reduced cell viability but had no effect on any other parameters. Conclusion: The LF-15 and T7 peptides may be appropriate candidates for use as novel pharmacotherapies due to thei

Brian G Oliver - One of the best experts on this subject based on the ideXlab platform.

  • Tumstatin fragment selectively inhibits neutrophil infiltration in experimental asthma exacerbation
    Clinical & Experimental Allergy, 2018
    Co-Authors: Gyde Nissen, Henrike Hollaender, Francesca Tang, Michael Wegmann, Lars Lunding, Christina Vock, Anna Bachmann, Solveig Lemmel, Rainer Bartels, Brian G Oliver
    Abstract:

    Background: Asthma is a chronic inflammatory disease with structural changes present. Burgess and colleagues recently found Tumstatin markedly reduced in adult asthmatic lung tissue compared with nonasthmatics. ECM fragments such as Tumstatin are named matrikines and act independently of the parent molecule. The role of Col IV matrikines in neutrophil inflammation (eg. exacerbation in asthma) has not been investigated to date. Severe adult asthma phenotypes are dominated by neutrophilic inflammation and show a high frequency of severe exacerbations. Objective: This study sought to investigate the role of a novel active region within Tumstatin (CP17) and its implication in neutrophil inflammatory responses related to asthma exacerbation. Methods: For reactive oxygen production, isolated neutrophils were preincubated with peptides or vehicle for 1 hour and stimulated (PMA). Luminescence signal was recorded (integration over 10 seconds) for 1.5 hours. Neutrophil migration was performed according to the SiMA protocol. Mice were sensitized to OVA/Alumn by intraperitoneal (i.p.) injections. Mice were then treated with CP17, vehicle (PBS) or scrambled peptide (SP17) after OVA exposure (days 27 and 28, polyI:C stimulation). All animals were killed on day 29 with lung function measurement, histology and lavage. Results: CP17 decreased total ROS production rate to 52.44% (0.5 mu mol/L, P <0.05 vs SP17), reduced the in vitro directionality (vs SP17, P = 1 x 10(-6)) and migration speed (5 mu mol/L, P = 1 x 10(-3)). In vivo application of CP17 decreased neutrophil inflammation similar to 1.8-fold (P <0.001 vs SP17) and reduced numbers of mucus-producing cells (-29%, P <0.05). Conclusion: CP17 reduced the ROS production rate, migrational speed and selectively inhibited neutrophil accumulation in the lung interstitium and lumen. Clinical relevance: CP17 may serve as a potential precursor for drug development to combat overwhelming neutrophil inflammation.

  • Tumstatin fragment selectively inhibits neutrophil infiltration in experimental asthma exacerbation.
    Clinical and experimental allergy : journal of the British Society for Allergy and Clinical Immunology, 2018
    Co-Authors: Gyde Nissen, Henrike Hollaender, Francesca Tang, Michael Wegmann, Lars Lunding, Christina Vock, Anna Bachmann, Solveig Lemmel, Rainer Bartels, Brian G Oliver
    Abstract:

    Background: Asthma is a chronic inflammatory disease with structural changes present. Burgess and colleagues recently found Tumstatin markedly reduced in adult asthmatic lung tissue compared with nonasthmatics. ECM fragments such as Tumstatin are named matrikines and act independently of the parent molecule. The role of Col IV matrikines in neutrophil inflammation (eg. exacerbation in asthma) has not been investigated to date. Severe adult asthma phenotypes are dominated by neutrophilic inflammation and show a high frequency of severe exacerbations. Objective: This study sought to investigate the role of a novel active region within Tumstatin (CP17) and its implication in neutrophil inflammatory responses related to asthma exacerbation. Methods: For reactive oxygen production, isolated neutrophils were preincubated with peptides or vehicle for 1 hour and stimulated (PMA). Luminescence signal was recorded (integration over 10 seconds) for 1.5 hours. Neutrophil migration was performed according to the SiMA protocol. Mice were sensitized to OVA/Alumn by intraperitoneal (i.p.) injections. Mice were then treated with CP17, vehicle (PBS) or scrambled peptide (SP17) after OVA exposure (days 27 and 28, polyI:C stimulation). All animals were killed on day 29 with lung function measurement, histology and lavage. Results: CP17 decreased total ROS production rate to 52.44% (0.5 mu mol/L, P

  • LF-15 & T7, synthetic peptides derived from Tumstatin, attenuate aspects of airway remodelling in a murine model of chronic OVA-induced allergic airway disease.
    PloS one, 2014
    Co-Authors: Karryn T Grafton, Janette K. Burgess, Judith L Black, Lyn M. Moir, Nicole G. Hansbro, Philip M. Hansbro, Brian G Oliver
    Abstract:

    Background Tumstatin is a segment of the collagen-IV protein that is markedly reduced in the airways of asthmatics. Tumstatin can play an important role in the development of airway remodelling associated with asthma due to its anti-angiogenic properties. This study assessed the anti-angiogenic properties of smaller peptides derived from Tumstatin, which contain the interface Tumstatin uses to interact with the αVβ3 integrin. Methods Primary human lung endothelial cells were exposed to the LF-15, T3 and T7 Tumstatin-derived peptides and assessed for cell viability and tube formation in vitro. The impact of the anti-angiogenic properties on airways hyperresponsiveness (AHR) was then examined using a murine model of chronic OVA-induced allergic airways disease. Results The LF-15 and T7 peptides significantly reduced endothelial cell viability and attenuated tube formation in vitro. Mice exposed to OVA+ LF-15 or OVA+T7 also had reduced total lung vascularity and AHR was attenuated compared to mice exposed to OVA alone. T3 peptides reduced cell viability but had no effect on any other parameters. Conclusion The LF-15 and T7 peptides may be appropriate candidates for use as novel pharmacotherapies due to their small size and anti-angiogenic properties observed in vitro and in vivo.

  • lf 15 t7 synthetic peptides derived from Tumstatin attenuate aspects of airway remodelling in a murine model of chronic ova induced allergic airway disease
    PLOS ONE, 2014
    Co-Authors: Karryn T Grafton, Janette K. Burgess, Judith L Black, Lyn M. Moir, Nicole G. Hansbro, Philip M. Hansbro, Brian G Oliver
    Abstract:

    Background Tumstatin is a segment of the collagen-IV protein that is markedly reduced in the airways of asthmatics. Tumstatin can play an important role in the development of airway remodelling associated with asthma due to its anti-angiogenic properties. This study assessed the anti-angiogenic properties of smaller peptides derived from Tumstatin, which contain the interface Tumstatin uses to interact with the αVβ3 integrin. Methods Primary human lung endothelial cells were exposed to the LF-15, T3 and T7 Tumstatin-derived peptides and assessed for cell viability and tube formation in vitro. The impact of the anti-angiogenic properties on airways hyperresponsiveness (AHR) was then examined using a murine model of chronic OVA-induced allergic airways disease. Results The LF-15 and T7 peptides significantly reduced endothelial cell viability and attenuated tube formation in vitro. Mice exposed to OVA+ LF-15 or OVA+T7 also had reduced total lung vascularity and AHR was attenuated compared to mice exposed to OVA alone. T3 peptides reduced cell viability but had no effect on any other parameters. Conclusion The LF-15 and T7 peptides may be appropriate candidates for use as novel pharmacotherapies due to their small size and anti-angiogenic properties observed in vitro and in vivo.

  • LF-15 & T7, Synthetic Peptides Derived from Tumstatin, Attenuate Aspects of Airway Remodelling in a Murine Model of Chronic OVA-Induced Allergic Airway Disease
    2014
    Co-Authors: Karryn T Grafton, Janette K. Burgess, Judith L Black, Lyn M. Moir, Nicole G. Hansbro, Philip M. Hansbro, Brian G Oliver
    Abstract:

    Background: Tumstatin is a segment of the collagen-IV protein that is markedly reduced in the airways of asthmatics. Tumstatin can play an important role in the development of airway remodelling associated with asthma due to its anti-angiogenic properties. This study assessed the anti-angiogenic properties of smaller peptides derived from Tumstatin, which contain the interface Tumstatin uses to interact with the aVb3 integrin. Methods: Primary human lung endothelial cells were exposed to the LF-15, T3 and T7 Tumstatin-derived peptides and assessed for cell viability and tube formation in vitro. The impact of the anti-angiogenic properties on airways hyperresponsiveness (AHR) was then examined using a murine model of chronic OVA-induced allergic airways disease. Results: The LF-15 and T7 peptides significantly reduced endothelial cell viability and attenuated tube formation in vitro. Mice exposed to OVA+ LF-15 or OVA+T7 also had reduced total lung vascularity and AHR was attenuated compared to mice exposed to OVA alone. T3 peptides reduced cell viability but had no effect on any other parameters. Conclusion: The LF-15 and T7 peptides may be appropriate candidates for use as novel pharmacotherapies due to thei

Yohei Maeshima - One of the best experts on this subject based on the ideXlab platform.

  • H: Tumstatin peptide, an inhibitor of angiogenesis, prevents glomerular hypertrophy in the early stage of diabetic nephropathy
    2016
    Co-Authors: Yoshihiko Yamamoto, Yohei Maeshima, Hiroyuki Kitayama, Shinji Kitamura, Yuki Takazawa, Hitoshi Sugiyama, Yasushi Yamasaki, Hirofumi Makino
    Abstract:

    In the early stage of diabetic nephropathy (one of the major microvascular complications of diabetes) glomer-ular hyperfiltration and hypertrophy are observed. It is clinically important to regulate glomerular hypertrophy for preventing glomerulosclerosis. The number of glomerular endothelial cells is known to be increased in diabetic nephropathy associated with enlarged glomer-ular tufts, suggesting that the mechanism is similar to that of angiogenesis. Tumstatin peptide is an angiogen-esis inhibitor derived from type IV collagen and inhibits in vivo neovascularization induced by vascular endothe-lial growth factor (VEGF), one of the mediators of glomerular hypertrophy in diabetic nephropathy. Here, we show the effect of Tumstatin peptide in inhibiting alterations in early diabetic nephropathy. Glomerula

  • identification of amino acids essential for the antiangiogenic activity of Tumstatin and its use in combination antitumor activity
    Proceedings of the National Academy of Sciences of the United States of America, 2008
    Co-Authors: Hans Petter Eikesdal, Yohei Maeshima, Hikaru Sugimoto, Gabriel Birrane, Vesselina G Cooke, Mark W Kieran, Raghu Kalluri
    Abstract:

    Tumstatin is an angiogenesis inhibitor that binds to αvβ3 integrin and suppresses tumor growth. Previous deletion mutagenesis studies identified a 25-aa fragment of Tumstatin (Tumstatin peptide) with in vitro antiangiogenic activity. Here, we demonstrate that systemic administration of this Tumstatin peptide inhibits tumor growth and angiogenesis. Site-directed mutagenesis identified amino acids L, V, and D as essential for the antiangiogenic activity of Tumstatin. The Tumstatin peptide binds to αvβ3 integrin on proliferating endothelial cells and localizes to select tumor endothelium in vivo. Using 3D molecular modeling, we identify a putative interaction interface for Tumstatin peptide on αvβ3 integrin. The antitumor activity of the Tumstatin peptide, in combination with bevacizumab (anti-VEGF antibody), displays significant improvement in efficacy against human renal cell carcinoma xenografts when compared with the single-agent use. Collectively, our results demonstrate that Tumstatin peptide binds specifically to the tumor endothelium, and its antiangiogenic action is mediated by αvβ3 integrin, and, in combination with an anti-VEGF antibody it exhibits enhanced tumor suppression of renal cell carcinoma.

  • Tumstatin Peptide, an Inhibitor of Angiogenesis, Prevents Glomerular Hypertrophy in the Early Stage of Diabetic Nephropathy
    Diabetes, 2004
    Co-Authors: Yoshihiko Yamamoto, Yohei Maeshima, Hiroyuki Kitayama, Shinji Kitamura, Yuki Takazawa, Hitoshi Sugiyama, Yasushi Yamasaki, Hirofumi Makino
    Abstract:

    In the early stage of diabetic nephropathy (one of the major microvascular complications of diabetes) glomerular hyperfiltration and hypertrophy are observed. It is clinically important to regulate glomerular hypertrophy for preventing glomerulosclerosis. The number of glomerular endothelial cells is known to be increased in diabetic nephropathy associated with enlarged glomerular tufts, suggesting that the mechanism is similar to that of angiogenesis. Tumstatin peptide is an angiogenesis inhibitor derived from type IV collagen and inhibits in vivo neovascularization induced by vascular endothelial growth factor (VEGF), one of the mediators of glomerular hypertrophy in diabetic nephropathy. Here, we show the effect of Tumstatin peptide in inhibiting alterations in early diabetic nephropathy. Glomerular hypertrophy, hyperfiltration, and albuminuria were suppressed by Tumstatin peptide (1 mg/kg) in streptozotocin-induced diabetic mice. Glomerular matrix expansion, the increase of total glomerular cell number and glomerular endothelial cells (CD31 positive), and monocyte/macrophage accumulation was inhibited by Tumstatin peptide. Increase in renal expression of VEGF, flk-1, and angiopoietin-2, an antagonist of angiopoietin-1, was inhibited by Tumstatin treatment in diabetic mice. Alteration of glomerular nephrin expression, a podocyte protein crucial for maintaining glomerular filtration barrier, was recovered by Tumstatin in diabetic mice. Taken together, these results demonstrate the potential use of antiangiogenic Tumstatin peptide as a novel therapeutic agent in early diabetic nephropathy.

  • physiological levels of Tumstatin a fragment of collagen iv α3 chain are generated by mmp 9 proteolysis and suppress angiogenesis via αvβ3 integrin
    Cancer Cell, 2003
    Co-Authors: Yuki Hamano, Yohei Maeshima, Julie C Lively, Richard O Hynes, Changqing Yang, Hikaru Sugimoto, Michael Zeisberg, Zena Werb, Akulapalli Sudhakar
    Abstract:

    We demonstrate a physiological role for Tumstatin, a cleavage fragment of the α3 chain of type IV collagen (Col IVα3), which is present in the circulation. Mice with a genetic deletion of Col IVα3 show accelerated tumor growth associated with enhanced pathological angiogenesis, while angiogenesis associated with development and tissue repair are unaffected. Supplementing Col IVα3-deficient mice with recombinant Tumstatin to a normal physiological concentration abolishes the increased rate of tumor growth. The suppressive effects of Tumstatin require αVβ3 integrin expressed on pathological, but not on physiological, angiogenic blood vessels. Mice deficient in matrix metalloproteinase-9, which cleaves Tumstatin efficiently from Col IVα3, have decreased circulating Tumstatin and accelerated growth of tumor. These results indicate that MMP-generated fragments of basement membrane collagen can have endogenous function as integrin-mediated suppressors of pathologic angiogenesis and tumor growth.

  • Tumstatin an endothelial cell specific inhibitor of protein synthesis
    Science, 2002
    Co-Authors: Yohei Maeshima, Akulapalli Sudhaka, Julie C Lively, Kohjiro Ueki, Surende Kharbanda, Nahum Sonenberg, Richard O Hynes, Roberta C Kah, Raghu Kalluri
    Abstract:

    Tumstatin is a 28-kilodalton fragment of type IV collagen that displays both anti-angiogenic and proapoptotic activity. Here we show that Tumstatin functions as an endothelial cell-specific inhibitor of protein synthesis. Through a requisite interaction with alphaVbeta3 integrin, Tumstatin inhibits activation of focal adhesion kinase (FAK), phosphatidylinositol 3-kinase (PI3-kinase), protein kinase B (PKB/Akt), and mammalian target of rapamycin (mTOR), and it prevents the dissociation of eukaryotic initiation factor 4E protein (eIF4E) from 4E-binding protein 1. These results establish a role for integrins in mediating cell-specific inhibition of cap-dependent protein synthesis and suggest a potential mechanism for Tumstatin's selective effects on endothelial cells.

In Sik Chung - One of the best experts on this subject based on the ideXlab platform.

  • Tumstatin induces apoptosis mediated by Fas signaling pathway in oral squamous cell carcinoma SCC-VII cells
    Oncology letters, 2015
    Co-Authors: Jeon Hwang-bo, Jong-hwa Park, In Sik Chung
    Abstract:

    Oral squamous cell carcinoma is a cancer originating in the tissues lining the mouth and lips. The present study investigated the effects of recombinant Tumstatin, an anti-angiogenic agent with distinct antitumor activity, on oral squamous cell carcinoma SCC-VII cells. Apoptosis was characterized by YO-PRO-1 staining, sub-G1 population, and DNA fragmentation analysis. Apoptotic mechanism of Tumstatin was also investigated. The antitumor activity of Tumstatin was further evaluated using an SCC-VII animal model. Recombinant Tumstatin was found to decrease the viability of SCC-VII cells in a dose-dependent manner. The number of cells stained with the apoptotic marker YO-PRO-1, the sub-G1 cell population and the level of apoptotic DNA fragmentation increased in the SCC-VII cells following treatment with recombinant Tumstatin. In addition, recombinant Tumstatin treatment increased the expression of the Fas gene at the transcript and protein levels, and the inhibition of cell viability by recombinant Tumstatin was suppressed by a neutralizing anti-Fas antibody. Furthermore, treatment with recombinant Tumstatin decreased the volume and weight of tumors in C3H/HeJ mice implanted with SCC-VII cells. In conclusion, the results indicated that Tumstatin induced apoptosis that is mediated by the Fas signaling pathway in SCC-VII cells and inhibited tumor growth in an SCC-VII animal model.

  • Peritumor injections of purified Tumstatin delay tumor growth and lymphatic metastasis in an orthotopic oral squamous cell carcinoma model.
    Oral oncology, 2008
    Co-Authors: In Sik Chung, Young-ik Son, Chung-hwan Baek, Jae Keun Cho, Han-sin Jeong
    Abstract:

    Tumstatin - non-collagenous (NC1) domain of the alpha 3 chain of type IV collagen - is a potent inhibitor of tumor angiogenesis. Successful tumor inhibition has been reported in glioma, bronchopulmonary cancer and melanoma experimental model. In this study, the effects of Tumstatin, in vitro and in vivo, were investigated in an oral cancer model. Recombinant human Tumstatin proteins were obtained by the transformation of Tn 5B1-4 cells, transfected with a plasmid containing Tumstatin cDNA using the lipofection method, as previously described. Tumstatin inhibited the proliferation of human umbilical vascular endothelial cells in a dose dependent manner in a proliferation assay. For the in vivo analysis, we established an orthotopic oral squamous cell carcinoma (AT-84 cells) animal (C3H/He) model. In this animal model, the in vivo inhibitory effects of Tumstatin on the tumor growth and on the metastasis of tumors were demonstrated. However, the tumors did not show complete remission. Immunostaining of the tumor microvessels (CD-31/PECAM) revealed that the density of tumor microvessels was significantly decreased in the Tumstatin treated primary tumors. The results demonstrated that Tumstatin delayed the tumor growth and the metastasis of oral squamous cell carcinomas. However, Tumstatin alone failed to achieve tumor regression. Therefore, Tumstatin might have an adjuvant role in the treatment of oral cancers, in combination with the conventional therapy.

  • Functional expression of recombinant canstatin in stably transformed Drosophila melanogaster S2 cells.
    Protein Expression and Purification, 2006
    Co-Authors: Hwang-bo Jeon, Bong-hee Sohn, In Sik Chung
    Abstract:

    Recombinant Tumstatin was expressed in stably transformed Drosophila melanogaster S2 cells and secreted into the medium with a molecular size of 29 kDa. Recombinant endostatin was also purified to homogeneity using a simple one-step Ni2+ affinity fractionation. Purified recombinant Tumstatin inhibited endothelial cell proliferation in a dose-dependent manner. The concentration at half-maximum inhibition for recombinant Tumstatin was approx. 0.7 μg ml−1. A maximum production of 4.6 μg recombinant Tumstatin (107 cells)−1 was obtained in a T-flask culture of S2 cells, 7 d after induction with 0.5 mM CuSO4.

  • Improved production of recombinant Tumstatin in stably transformed Trichoplusia ni BTI Tn 5B1-4 cells.
    Protein expression and purification, 2004
    Co-Authors: Kyung Hwa Chang, Hee Kyoung Jeon, Jong Min Lee, In Sik Chung
    Abstract:

    Abstract We describe the expression and in vitro activity of recombinant Tumstatin from stably transformed Trichoplusia ni BTI Tn 5B1-4 cells. Recombinant Tumstatin was secreted into a culture medium with a molecular weight of 29 kDa. Recombinant Tumstatin was also purified to homogeneity using a simple one-step Ni 2+ affinity fractionation. Purified recombinant Tumstatin inhibited endothelial cell proliferation in a dose-dependent manner. The concentration at half-maximum inhibition (ED 50 ) for recombinant Tumstatin expressed in stably transformed Tn 5B1-4 cells was approximately 0.76 μg/ml. A maximum production level of 4.0 mg/l recombinant Tumstatin was obtained in a T-flask culture of Tn 5B1-4 cells, 6 days after cultivation. We also investigated the individual effects of both dimethyl sulfoxide (DMSO) and sodium butyrate on recombinant Tumstatin production in stably transformed Tn 5B1-4 cells. Supplementing cultures with DMSO and sodium butyrate separately increased recombinant Tumstatin production in stably transformed Tn 5B1-4 cells by 117 and 32%, respectively.

  • Functional expression of recombinant Tumstatin in stably transformed Drosophila melanogaster S2 cells
    Biotechnology Letters, 2003
    Co-Authors: Hee Kyoung Jeon, Kyung Hwa Chang, In Sik Chung
    Abstract:

    Recombinant Tumstatin was expressed in stably transformed Drosophila melanogaster S2 cells and secreted into the medium with a molecular size of 29 kDa. Recombinant endostatin was also purified to homogeneity using a simple one-step Ni^2+ affinity fractionation. Purified recombinant Tumstatin inhibited endothelial cell proliferation in a dose-dependent manner. The concentration at half-maximum inhibition for recombinant Tumstatin was approx. 0.7 μg ml^−1. A maximum production of 4.6 μg recombinant Tumstatin (10^7 cells)^−1 was obtained in a T-flask culture of S2 cells, 7 d after induction with 0.5 mM CuSO_4.