Network


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

Hotspot


Dive into the research topics where Brian K. Stansfield is active.

Publication


Featured researches published by Brian K. Stansfield.


Circulation | 2014

Neurofibromin-deficient myeloid cells are critical mediators of aneurysm formation in vivo

Fang Li; Brandon D. Downing; Lucy C. Smiley; Julie A. Mund; Matthew R. DiStasi; Waylan K. Bessler; Kara N. Sarchet; Daniel M. Hinds; Lisa M. Kamendulis; Cynthia M. Hingtgen; Jamie Case; D. Wade Clapp; Simon J. Conway; Brian K. Stansfield; David A. Ingram

Background— Neurofibromatosis type 1 (NF1) is a genetic disorder resulting from mutations in the NF1 tumor suppressor gene. Neurofibromin, the protein product of NF1, functions as a negative regulator of Ras activity in circulating hematopoietic and vascular wall cells, which are critical for maintaining vessel wall homeostasis. NF1 patients have evidence of chronic inflammation resulting in the development of premature cardiovascular disease, including arterial aneurysms, which may manifest as sudden death. However, the molecular pathogenesis of NF1 aneurysm formation is unknown. Method and Results— With the use of an angiotensin II–induced aneurysm model, we demonstrate that heterozygous inactivation of Nf1 (Nf1+/–) enhanced aneurysm formation with myeloid cell infiltration and increased oxidative stress in the vessel wall. Using lineage-restricted transgenic mice, we show that loss of a single Nf1 allele in myeloid cells is sufficient to recapitulate the Nf1+/– aneurysm phenotype in vivo. Finally, oral administration of simvastatin or the antioxidant apocynin reduced aneurysm formation in Nf1+/– mice. Conclusion— These data provide genetic and pharmacological evidence that Nf1+/– myeloid cells are the cellular triggers for aneurysm formation in a novel model of NF1 vasculopathy and provide a potential therapeutic target.


American Journal of Pathology | 2014

Ras-Mek-Erk Signaling Regulates Nf1 Heterozygous Neointima Formation

Brian K. Stansfield; Waylan K. Bessler; Raghuveer Singh Mali; Julie A. Mund; Brandon D. Downing; Reuben Kapur; David A. Ingram

Neurofibromatosis type 1 (NF1) results from mutations in the NF1 tumor-suppressor gene, which encodes neurofibromin, a negative regulator of diverse Ras signaling cascades. Arterial stenosis is a nonneoplastic manifestation of NF1 that predisposes some patients to debilitating morbidity and sudden death. Recent murine studies demonstrate that Nf1 heterozygosity (Nf1(+/-)) in monocytes/macrophages significantly enhances intimal proliferation after arterial injury. However, the downstream Ras effector pathway responsible for this phenotype is unknown. Based on inxa0vitro assays demonstrating enhanced extracellular signal-related kinase (Erk) signaling in Nf1(+/-) macrophages and vascular smooth muscle cells and inxa0vivo evidence of Erk amplification without alteration of phosphatidylinositol 3-kinase signaling in Nf1(+/-) neointimas, we tested the hypothesis that Ras-Erk signaling regulates intimal proliferation in a murine model of NF1 arterial stenosis. By using a well-established inxa0vivo model of inflammatory cell migration and standard cell culture, neurofibromin-deficient macrophages demonstrate enhanced sensitivity to growth factor stimulation inxa0vivo and inxa0vitro, which is significantly diminished in the presence of PD0325901, a specific inhibitor of Ras-Erk signaling in phase 2 clinical trials for cancer. After carotid artery injury, Nf1(+/-) mice demonstrated increased intimal proliferation compared with wild-type mice. Daily administration of PD0325901 significantly reduced Nf1(+/-) neointima formation to levels of wild-type mice. These studies identify the Ras-Erk pathway in neurofibromin-deficient macrophages as the aberrant pathway responsible for enhanced neointima formation.


Human Molecular Genetics | 2013

Heterozygous Inactivation of the Nf1 Gene in Myeloid Cells Enhances Neointima Formation via a Rosuvastatin-Sensitive Cellular Pathway

Brian K. Stansfield; Waylan K. Bessler; Raghuveer Singh Mali; Julie A. Mund; Brandon D. Downing; Fang Li; Kara N. Sarchet; Matthew R. DiStasi; Simon J. Conway; Reuben Kapur; David A. Ingram

Mutations in the NF1 tumor suppressor gene cause Neurofibromatosis type 1 (NF1). Neurofibromin, the protein product of NF1, functions as a negative regulator of Ras activity. Some NF1 patients develop cardiovascular disease, which represents an underrecognized disease complication and contributes to excess morbidity and mortality. Specifically, NF1 patients develop arterial occlusion resulting in tissue ischemia and sudden death. Murine studies demonstrate that heterozygous inactivation of Nf1 (Nf1(+/-)) in bone marrow cells enhances neointima formation following arterial injury. Macrophages infiltrate Nf1(+/-) neointimas, and NF1 patients have increased circulating inflammatory monocytes in their peripheral blood. Therefore, we tested the hypothesis that heterozygous inactivation of Nf1 in myeloid cells is sufficient for neointima formation. Specific ablation of a single copy of the Nf1 gene in myeloid cells alone mobilizes a discrete pro-inflammatory murine monocyte population via a cell autonomous and gene-dosage dependent mechanism. Furthermore, lineage-restricted heterozygous inactivation of Nf1 in myeloid cells is sufficient to reproduce the enhanced neointima formation observed in Nf1(+/-) mice when compared with wild-type controls, and homozygous inactivation of Nf1 in myeloid cells amplified the degree of arterial stenosis after arterial injury. Treatment of Nf1(+/-) mice with rosuvastatin, a stain with anti-inflammatory properties, significantly reduced neointima formation when compared with control. These studies identify neurofibromin-deficient myeloid cells as critical cellular effectors of Nf1(+/-) neointima formation and propose a potential therapeutic for NF1 cardiovascular disease.


Human Molecular Genetics | 2016

Nf1+/− monocytes/macrophages induce neointima formation via CCR2 activation

Waylan K. Bessler; Grace Kim; Farlyn Z. Hudson; Julie A. Mund; Raghuveer Singh Mali; Keshav Menon; Reuben Kapur; D. Wade Clapp; David A. Ingram; Brian K. Stansfield

Persons with neurofibromatosis type 1 (NF1) have a predisposition for premature and severe arterial stenosis. Mutations in the NF1 gene result in decreased expression of neurofibromin, a negative regulator of p21(Ras), and increases Ras signaling. Heterozygous Nf1 (Nf1(+/-)) mice develop a marked arterial stenosis characterized by proliferating smooth muscle cells (SMCs) and a predominance of infiltrating macrophages, which closely resembles arterial lesions from NF1 patients. Interestingly, lineage-restricted inactivation of a single Nf1 allele in monocytes/macrophages is sufficient to recapitulate the phenotype observed in Nf1(+/-) mice and to mobilize proinflammatory CCR2+ monocytes into the peripheral blood. Therefore, we hypothesized that CCR2 receptor activation by its primary ligand monocyte chemotactic protein-1 (MCP-1) is critical for monocyte infiltration into the arterial wall and neointima formation in Nf1(+/-) mice. MCP-1 induces a dose-responsive increase in Nf1(+/-) macrophage migration and proliferation that corresponds with activation of multiple Ras kinases. In addition, Nf1(+/-) SMCs, which express CCR2, demonstrate an enhanced proliferative response to MCP-1 when compared with WT SMCs. To interrogate the role of CCR2 activation on Nf1(+/-) neointima formation, we induced neointima formation by carotid artery ligation in Nf1(+/-) and WT mice with genetic deletion of either MCP1 or CCR2. Loss of MCP-1 or CCR2 expression effectively inhibited Nf1(+/-) neointima formation and reduced macrophage content in the arterial wall. Finally, administration of a CCR2 antagonist significantly reduced Nf1(+/-) neointima formation. These studies identify MCP-1 as a potent chemokine for Nf1(+/-) monocytes/macrophages and CCR2 as a viable therapeutic target for NF1 arterial stenosis.


Free Radical Biology and Medicine | 2016

Neurofibromin is a novel regulator of Ras-induced reactive oxygen species production in mice and humans

Waylan K. Bessler; Farlyn Z. Hudson; Hanfang Zhang; Valerie Harris; Yusi Wang; Julie A. Mund; Brandon D. Downing; David A. Ingram; Jamie Case; David Fulton; Brian K. Stansfield

Neurofibromatosis type 1 (NF1) predisposes individuals to early and debilitating cardiovascular disease. Loss of function mutations in the NF1 tumor suppressor gene, which encodes the protein neurofibromin, leads to accelerated p21(Ras) activity and phosphorylation of multiple downstream kinases, including Erk and Akt. Nf1 heterozygous (Nf1(+/-)) mice develop a robust neointima that mimics human disease. Monocytes/macrophages play a central role in NF1 arterial stenosis as Nf1 mutations in myeloid cells alone are sufficient to reproduce the enhanced neointima observed in Nf1(+/-) mice. Though the molecular mechanisms underlying NF1 arterial stenosis remain elusive, macrophages are important producers of reactive oxygen species (ROS) and Ras activity directly regulates ROS production. Here, we use compound mutant and lineage-restricted mice to demonstrate that Nf1(+/-) macrophages produce excessive ROS, which enhance Nf1(+/-) smooth muscle cell proliferation in vitro and in vivo. Further, use of a specific NADPH oxidase-2 inhibitor to limit ROS production prevents neointima formation in Nf1(+/-) mice. Finally, mononuclear cells from asymptomatic NF1 patients have increased oxidative DNA damage, an indicator of chronic exposure to oxidative stress. These data provide genetic and pharmacologic evidence that excessive exposure to oxidant species underlie NF1 arterial stenosis and provide a platform for designing novels therapies and interventions.


Archive | 2012

Molecular Basis of Cardiovascular Abnormalities in NF1

Brian K. Stansfield; David A. Ingram; Simon J. Conway; Jan M. Friedman

Congenital heart defects are uncommon among people with NF1, but pulmonic stenosis and coarctation of the aorta appear to occur more often than expected. Double outlet right ventricle (the characteristic cardiac defect in Nf1 −/− mouse models) and other complex cardiac malformations are rare in people with NF1.


PMC | 2016

Nf1+/- monocytes/macrophages induce neointima formation via CCR2 activation

Waylan K. Bessler; Grace Kim; Farlyn Z. Hudson; Julie A. Mund; Raghuveer Singh Mali; Keshav Menon; Reuben Kapur; D. Wade Clapp; David A. Ingram; Brian K. Stansfield


PMC | 2015

Clinical significance of monocyte heterogeneity

Brian K. Stansfield; David A. Ingram


PMC | 2014

Neurofibromin Deficient Myeloid Cells are Critical Mediators of Aneurysm Formation In Vivo

Fang Li; Brandon D. Downing; Lucy C. Smiley; Julie A. Mund; Matthew R. DiStasi; Waylan K. Bessler; Kara N. Sarchet; Daniel M. Hinds; Lisa M. Kamendulis; Cynthia M. Hingtgen; Jamie Case; D. Wade Clapp; Simon J. Conway; Brian K. Stansfield; David A. Ingram

Collaboration


Dive into the Brian K. Stansfield's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Farlyn Z. Hudson

Georgia Regents University

View shared research outputs
Researchain Logo
Decentralizing Knowledge