Network


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

Hotspot


Dive into the research topics where Juliet G. Carbon is active.

Publication


Featured researches published by Juliet G. Carbon.


Clinical Cancer Research | 2007

Monitoring Response to Anticancer Therapy by Targeting Microbubbles to Tumor Vasculature

Grzegorz Korpanty; Juliet G. Carbon; Paul A. Grayburn; Jason B. Fleming; Rolf A. Brekken

Purpose: New strategies to detect tumor angiogenesis and monitor response of tumor vasculature to therapy are needed. Contrast ultrasound imaging using microbubbles targeted to tumor endothelium offers a noninvasive method for monitoring and quantifying vascular effects of antitumor therapy. We investigated the use of targeted microbubbles to follow vascular response of therapy in a mouse model of pancreatic adenocarcinoma. Experimental Design: Microbubbles conjugated to monoclonal antibodies were used to image and quantify vascular effects of two different antitumor therapies in s.c. and orthotopic pancreatic tumors in mice. Tumor-bearing mice were treated with anti-vascular endothelial growth factor (VEGF) monoclonal antibodies and/or gemcitabine, and the localization of microbubbles to endoglin (CD105), VEGF receptor 2 (VEGFR2), or VEGF-activated blood vessels (the VEGF-VEGFR complex) was monitored by contrast ultrasound. Results: Targeted microbubbles showed significant enhancement of tumor vasculature when compared with untargeted or control IgG–targeted microbubbles. Video intensity from targeted microbubbles correlated with the level of expression of the target (CD105, VEGFR2, or the VEGF-VEGFR complex) and with microvessel density in tumors under antiangiogenic or cytotoxic therapy. Conclusions: We conclude that targeted microbubbles represent a novel and attractive tool for noninvasive, vascular-targeted molecular imaging of tumor angiogenesis and for monitoring vascular effects specific to antitumor therapy in vivo.


Cancer Research | 2010

Smac mimetic increases chemotherapy response and improves survival in mice with pancreatic cancer.

Sean P. Dineen; Christina L. Roland; Rachel Greer; Juliet G. Carbon; Jason E. Toombs; Puja Gupta; Nabeel Bardeesy; Haizhou Sun; Noelle S. Williams; John D. Minna; Rolf A. Brekken

Failure of chemotherapy in the treatment of pancreatic cancer is often due to resistance to therapy-induced apoptosis. A major mechanism for such resistance is the expression and activity of inhibitors of apoptosis proteins (IAP). Smac (second mitochondria-derived activator of caspase) is a mitochondrial protein that inhibits IAPs. We show that JP1201, a Smac mimetic, is a potent enhancer of chemotherapy in robust mouse models of pancreatic cancer. Combination of JP1201 with gemcitabine reduced primary and metastatic tumor burden in orthotopic xenograft and syngenic tumor models, induced regression of established tumors, and prolonged survival in xenograft and transgenic models of pancreatic cancer. The effect of JP1201 was phenocopied by XIAP small interfering RNA in vitro and correlated with elevated levels of tumor necrosis factor alpha protein in vivo. The continued development of JP1201 and other strategies designed to enhance therapy-induced apoptosis in pancreatic cancer is warranted.


Disease Models & Mechanisms | 2010

Lack of host SPARC enhances vascular function and tumor spread in an orthotopic murine model of pancreatic carcinoma

Shanna A. Arnold; Lee B. Rivera; Andrew F. Miller; Juliet G. Carbon; Sean P. Dineen; Yang Xie; Diego H. Castrillon; E. Helene Sage; Pauli Puolakkainen; Amy D. Bradshaw; Rolf A. Brekken

SUMMARY Utilizing subcutaneous tumor models, we previously validated SPARC (secreted protein acidic and rich in cysteine) as a key component of the stromal response, where it regulated tumor size, angiogenesis and extracellular matrix deposition. In the present study, we demonstrate that pancreatic tumors grown orthotopically in Sparc-null (Sparc−/−) mice are more metastatic than tumors grown in wild-type (Sparc+/+) littermates. Tumors grown in Sparc−/− mice display reduced deposition of fibrillar collagens I and III, basement membrane collagen IV and the collagen-associated proteoglycan decorin. In addition, microvessel density and pericyte recruitment are reduced in tumors grown in the absence of host SPARC. However, tumors from Sparc−/− mice display increased permeability and perfusion, and a subsequent decrease in hypoxia. Finally, we found that tumors grown in the absence of host SPARC exhibit an increase in alternatively activated macrophages. These results suggest that increased tumor burden in the absence of host SPARC is a consequence of reduced collagen deposition, a disrupted vascular basement membrane, enhanced vascular function and an immune-tolerant, pro-metastatic microenvironment.


BMC Cancer | 2008

The Adnectin CT-322 is a novel VEGF receptor 2 inhibitor that decreases tumor burden in an orthotopic mouse model of pancreatic cancer

Sean P. Dineen; Laura A. Sullivan; Adam W. Beck; Andrew F. Miller; Juliet G. Carbon; Roni Mamluk; Henry Wong; Rolf A. Brekken

BackgroundPancreatic cancer continues to have a 5-year survival of less than 5%. Therefore, more effective therapies are necessary to improve prognosis in this disease. Angiogenesis is required for tumor growth, and subsequently, mediators of angiogenesis are attractive targets for therapy. Vascular endothelial growth factor (VEGF) is a well-characterized mediator of tumor angiogenesis that functions primarily by binding and activating VEGF receptor 2 (VEGFR2). In this study, we evaluate the use of CT-322, a novel biologic (Adnectin). This small protein is based on a human fibronectin domain and has beneficial properties in that it is fully human, stable, and is produced in bacteria. CT-322 binds to and inhibits activation of VEGFR2.MethodsThe efficacy of CT-322 was evaluated in vivo using two orthotopic pancreatic tumor models. The first model was a human tumor xenograft where MiaPaCa-2 cells were injected into the tail of the pancreas of nude mice. The second model was a syngeneic tumor using Pan02 cells injected into pancreas of C57BL/6J mice. In both models, therapy was initiated once primary tumors were established. Mice bearing MiaPaCa-2 tumors were treated with vehicle or CT-322 alone. Gemcitabine alone or in combination with CT-322 was added to the treatment regimen of mice bearing Pan02 tumors. Therapy was given twice a week for six weeks, after which the animals were sacrificed and evaluated (grossly and histologically) for primary and metastatic tumor burden. Primary tumors were also evaluated by immunohistochemistry for the level of apoptosis (TUNEL), microvessel density (MECA-32), and VEGF-activated blood vessels (Gv39M).ResultsTreatment with CT-322 was effective at preventing pancreatic tumor growth and metastasis in orthotopic xenograft and syngeneic models of pancreatic cancer. Additionally, CT-322 treatment increased apoptosis, reduced microvessel density and reduced the number of VEGF-activated blood vessels in tumors. Finally, CT-322, in combination with gemcitabine was safe and effective at controlling the growth of syngeneic pancreatic tumors in immunocompetent mice.ConclusionWe conclude that CT-322 is an effective anti-VEGFR2 agent and that further investigation of CT-322 for the treatment of pancreatic cancer is warranted.


Journal of Cellular Biochemistry | 2003

SPARC regulates cell cycle progression in mesangial cells via its inhibition of IGF‐dependent signaling

Aleksandar Francki; Kouros Motamed; Timothy D. McClure; Marcus Kaya; Carrie Murri; David J. Blake; Juliet G. Carbon; E. Helene Sage

Glomerular mesangial cells both synthesize and respond to insulin‐like growth factor‐1 (IGF‐1). Increased activity of the IGF signaling pathway has been implicated as a major contributor to renal enlargement and subsequent development of diabetic nephropathy. Secreted protein acidic and rich in cysteine (SPARC), a matricellular protein, has been shown to modulate the interaction of cells with growth factors and extracellular matrix. We have reported that primary glomerular mesangial cells derived from SPARC‐null mice exhibit an accelerated rate of proliferation and produce substantially decreased levels of transforming growth factor β1 (TGF‐β1) in comparison to their wild‐type counterparts (Francki et al. [ 1999 ] J. Biol. Chem. 274: 32145–32152). Herein we present evidence that SPARC modulates IGF‐dependent signaling in glomerular mesangial cells. SPARC‐null mesangial cells produce increased amounts of IGF‐1 and ‐2, as well as IGF‐1 receptor (IGF‐1R) in comparison to wild‐type cells. Addition of recombinant SPARC to SPARC‐null cells inhibited IGF‐1‐stimulated mitogen activated protein kinase (MAPK) activation and DNA synthesis. We also show that the observed accelerated rate of basal and IGF‐1‐stimulated proliferation in mesangial cells derived from SPARC‐null animals is due, at least in part, to markedly diminished levels of cyclin D1 and the cyclin‐dependent kinase (cdk) inhibitors p21 and p27. Since expression of SPARC in the glomerulus is especially prominent during renal injury, our findings substantiate previous claims that SPARC is involved in glomerular remodeling and repair, a process commonly associated with mesangioproliferative glomerulonephritis and diabetic nephropathy.


Wound Repair and Regeneration | 2001

Increased fibrovascular invasion of subcutaneous polyvinyl alcohol sponges in SPARC-null mice

Amy D. Bradshaw; May J. Reed; Juliet G. Carbon; Emmett Pinney; Rolf A. Brekken; E. Helene Sage

The expression of SPARC (secreted protein acidic and rich in cysteine/osteonectin/BM‐40) is elevated in endothelial cells participating in angiogenesis in vitro and in vivo. SPARC acts on endothelial cells to elicit changes in cell shape and to inhibit cell cycle progression. In addition, SPARC binds to and diminishes the mitotic activity of vascular endothelial growth factor. To determine the effect(s) of SPARC on angiogenic responses in vivo, we implanted polyvinyl alcohol sponges subcutaneously into wild‐type and SPARC‐null mice. On days 12 and 20 following implantation, SPARC‐null mice showed increased cellular invasion of the sponges in comparison to wild‐type mice. Areas of the sponge with the highest cell density exhibited the highest numbers of vascular profiles in both wild‐type and SPARC‐null animals. The endothelial component of the vessels was substantiated by immunoreactivity with three different markers specific for endothelial cells. Although sponges from SPARC‐null relative to wild‐type mice were populated by significantly more cells and blood vessels, an increase in the ratio of vascular to nonvascular cells was not apparent. No differences in the percentage of proliferating cells within the sponge were detected between wild‐type and SPARC‐null sections. However, elevated levels of vascular endothelial growth factor were associated with sponges from SPARC‐null versus wild‐type mice. An increase in vascular endothelial growth factor production was also observed in SPARC‐null primary dermal fibroblasts relative to those of wild‐type cells. In conclusion, we have shown that the fibrovascular invasion of polyvinyl alcohol sponges is enhanced in mice lacking SPARC, and we propose that increased levels of vascular endothelial growth factor account, at least in part, for this response.


Journal of Histochemistry and Cytochemistry | 2004

Functional Analysis of the Matricellular Protein SPARC with Novel Monoclonal Antibodies

Mariya T. Sweetwyne; Rolf A. Brekken; Gail Workman; Amy D. Bradshaw; Juliet G. Carbon; Anthony W. Siadak; Carrie Murri; E. Helene Sage

SPARC (osteonectin, BM-40) is a matricellular glycoprotein that is expressed in many embryogenic and adult tissues undergoing remodeling or repair. SPARC modulates cellular interaction with the extracellular matrix (ECM), inhibits cell adhesion and proliferation, and regulates growth factor activity. To explore further the function and activity of this protein in tissue homeostasis, we have developed several monoclonal antibodies (MAbs) that recognize distinct epitopes on SPARC. The MAbs bind to SPARC with high affinity and identify SPARC by ELISA, Western blotting, immunoprecipitation, immunocytochemistry, and/or immunohistochemistry. The MAbs were also characterized in functional assays for potential alteration of SPARC activity. SPARC binds to collagen I and laminin-1 through an epitope defined by MAb 293; this epitope is not involved in the binding of SPARC to collagen III. The other MAbs did not interfere with the binding of SPARC to collagen I or III or laminin-1. Inhibition of the anti-adhesive effect of SPARC on endothelial cells by MAb 236 was also observed. Functional analysis of SPARC in the presence of these novel MAbs now confirms that the activities ascribed to this matricellular protein can be assigned to discrete subdomains.


Journal of Histochemistry and Cytochemistry | 2004

Expression and Characterization of Murine Hevin (SC1), a Member of the SPARC Family of Matricellular Proteins

Rolf A. Brekken; Millicent M. Sullivan; Gail Workman; Amy D. Bradshaw; Juliet G. Carbon; Anthony W. Siadak; Carrie Murri; Paul E. Framson; E. Helene Sage

Hevin, also known as SC1, MAST 9, SPARC-like 1, RAGS1 and ECM2, is a member of the SPARC-related family of matricellular proteins. Mouse hevin is 53% identical to mouse SPARC, and both proteins share a follistatin-like module and an extracellular Ca2+-binding (E-C) domain. SPARC functions as a modulator of cell-matrix interactions, a regulator of growth factor activity, a de-adhesive protein, and a cell cycle inhibitor. Although the functions of mouse hevin are unknown, its human orthologue has been shown to be de-adhesive for endothelial cells. We now report the production of recombinant mouse hevin in insect cells through the use of a baculoviral expression system and its purification by anion-exchange, size-exclusion chromatography, and isoelectric focusing. Furthermore, we have produced rat anti-hevin monoclonal antibodies (MAbs) that have been characterized by indirect and capture ELISAs, immunoblotting, immunoprecipitation, and immunohistochemistry (IHC). Recombinant hevin, present as a soluble factor or bound to tissue-culture plastic, inhibited the spreading of bovine aortic endothelial cells in vitro. IHC analysis of hevin in normal human and mouse tissues revealed a limited expression pattern in many tissues, with particularly dominant staining in dermis, ducts, vasculature, muscle, and brain. In lung and pancreatic tumor xenografts, we found distinct reactivity with MAbs that were selective for stromal cells, tumor cells, and/or endothelial cells. Although similar to SPARC in its anti-adhesive activities, hevin nevertheless exhibits a distinctive histological distribution that, in certain invasive tumors, is associated with desmoplasia.


PLOS ONE | 2012

Losartan Slows Pancreatic Tumor Progression and Extends Survival of SPARC-Null Mice by Abrogating Aberrant TGFβ Activation

Shanna A. Arnold; Lee B. Rivera; Juliet G. Carbon; Jason E. Toombs; Chi-Lun Chang; Amy D. Bradshaw; Rolf A. Brekken

Pancreatic adenocarcinoma, a desmoplastic disease, is the fourth leading cause of cancer-related death in the Western world due, in large part, to locally invasive primary tumor growth and ensuing metastasis. SPARC is a matricellular protein that governs extracellular matrix (ECM) deposition and maturation during tissue remodeling, particularly, during wound healing and tumorigenesis. In the present study, we sought to determine the mechanism by which lack of host SPARC alters the tumor microenvironment and enhances invasion and metastasis of an orthotopic model of pancreatic cancer. We identified that levels of active TGFβ1 were increased significantly in tumors grown in SPARC-null mice. TGFβ1 contributes to many aspects of tumor development including metastasis, endothelial cell permeability, inflammation and fibrosis, all of which are altered in the absence of stromal-derived SPARC. Given these results, we performed a survival study to assess the contribution of increased TGFβ1 activity to tumor progression in SPARC-null mice using losartan, an angiotensin II type 1 receptor antagonist that diminishes TGFβ1 expression and activation in vivo. Tumors grown in SPARC-null mice progressed more quickly than those grown in wild-type littermates leading to a significant reduction in median survival. However, median survival of SPARC-null animals treated with losartan was extended to that of losartan-treated wild-type controls. In addition, losartan abrogated TGFβ induced gene expression, reduced local invasion and metastasis, decreased vascular permeability and altered the immune profile of tumors grown in SPARC-null mice. These data support the concept that aberrant TGFβ1-activation in the absence of host SPARC contributes significantly to tumor progression and suggests that SPARC, by controlling ECM deposition and maturation, can regulate TGFβ availability and activation.


Cancer Research | 2014

Collagen Signaling Enhances Tumor Progression after Anti-VEGF Therapy in a Murine Model of Pancreatic Ductal Adenocarcinoma

Kristina Y. Aguilera; Lee B. Rivera; Hoon Hur; Juliet G. Carbon; Jason E. Toombs; Courtney D. Goldstein; Michael T. Dellinger; Diego H. Castrillon; Rolf A. Brekken

There is growing evidence that antiangiogenic therapy stimulates cancer cell invasion and metastasis. However, the underlying molecular mechanisms responsible for these changes have not been fully defined. Here, we report that anti-VEGF therapy promotes local invasion and metastasis by inducing collagen signaling in cancer cells. We show that chronic VEGF inhibition in a genetically engineered mouse model of pancreatic ductal adenocarcinoma (PDA) induces hypoxia, a less differentiated mesenchymal-like tumor cell phenotype, TGF-β expression, and collagen deposition and signaling. In addition, we show that collagen signaling is critical for protumorigenic activity of TGF-β in vitro. To further model the impact of collagen signaling in tumors, we evaluated PDA in mice lacking Sparc, a protein that reduces collagen binding to cell surface receptors. Importantly, we show that loss of Sparc increases collagen signaling and tumor progression. Together, these findings suggest that collagen actively promotes PDA spread and that enhanced disease progression associated with anti-VEGF therapy can arise from elevated extracellular matrix-mediated signaling.

Collaboration


Dive into the Juliet G. Carbon's collaboration.

Top Co-Authors

Avatar

Rolf A. Brekken

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jason E. Toombs

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Amy D. Bradshaw

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Sean P. Dineen

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

E. Helene Sage

Benaroya Research Institute

View shared research outputs
Top Co-Authors

Avatar

Lee B. Rivera

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Shanna A. Arnold

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Andrew F. Miller

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Christina L. Roland

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge