Network


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

Hotspot


Dive into the research topics where Seth B. Furgeson is active.

Publication


Featured researches published by Seth B. Furgeson.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2011

SDF-1α Induction in Mature Smooth Muscle Cells by Inactivation of PTEN Is a Critical Mediator of Exacerbated Injury-Induced Neointima Formation

Raphael A. Nemenoff; Henrick Horita; Allison C. Ostriker; Seth B. Furgeson; Peter Simpson; Vicki VanPutten; Joseph T. Crossno; Stefan Offermanns; Mary C.M. Weiser-Evans

Objective—PTEN inactivation selectively in smooth muscle cells (SMC) initiates multiple downstream events driving neointima formation, including SMC cytokine/chemokine production, in particular stromal cell-derived factor-1&agr; (SDF-1&agr;). We investigated the effects of SDF-1&agr; on resident SMC and bone marrow–derived cells and in mediating neointima formation. Methods and Results—Inducible, SMC-specific PTEN knockout mice (PTEN iKO) were bred to floxed-stop ROSA26-&bgr;-galactosidase (&bgr;Gal) mice to fate-map mature SMC in response to injury; mice received wild-type green fluorescent protein–labeled bone marrow to track recruitment. Following wire-induced femoral artery injury, &bgr;Gal(+) SMC accumulated in the intima and adventitia. Compared with wild-type, PTEN iKO mice exhibited massive neointima formation, increased replicating intimal and medial &bgr;Gal(+)SMC, and enhanced vascular recruitment of bone marrow cells following injury. Inhibiting SDF-1&agr; blocked these events and reversed enhanced neointima formation observed in PTEN iKO mice. Most recruited green fluorescent protein(+) cells stained positive for macrophage markers but not SMC markers. SMC-macrophage interactions resulted in a persistent SMC inflammatory phenotype that was dependent on SMC PTEN and SDF-1&agr; expression. Conclusion—Resident SMC play a multifaceted role in neointima formation by contributing the majority of neointimal cells, regulating recruitment of inflammatory cells, and contributing to adventitial remodeling. The SMC PTEN-SDF-1&agr; axis is a critical regulator of these events.


Circulation Research | 2008

Targeted Deletion of PTEN in Smooth Muscle Cells Results in Vascular Remodeling and Recruitment of Progenitor Cells Through Induction of Stromal Cell–Derived Factor-1α

Raphael A. Nemenoff; Peter Simpson; Seth B. Furgeson; Nihal Kaplan-Albuquerque; Joseph T. Crossno; Pamela J. Garl; James K. Cooper; Mary C.M. Weiser-Evans

We previously showed that changes in vascular smooth muscle cell (SMC) PTEN/Akt signaling following vascular injury are associated with increased SMC proliferation and neointima formation. In this report, we used a genetic model to deplete PTEN specifically in SMCs by crossing PTENLoxP/LoxP mice to mice expressing Cre recombinase under the control of the SM22&agr; promoter. PTEN was downregulated with increases in phosphorylated Akt in major vessels, hearts, and lungs of mutant mice. SMC PTEN depletion promoted widespread medial SMC hyperplasia, vascular remodeling, and histopathology consistent with pulmonary hypertension. Increased vascular deposition of the chemokine stromal cell–derived factor (SDF)-1&agr; and medial and intimal cells coexpressing SM-&agr;-actin and CXCR4, the SDF-1&agr; receptor, was detected in SMC PTEN-depleted mice. PTEN deficiency in cultured aortic SMCs induced autocrine growth through increased production of SDF-1&agr;. Blocking SDF-1&agr; attenuated autocrine growth and blocked growth of control SMCs induced by conditioned media from PTEN-deficient SMCs. In addition, SMC PTEN deficiency enhanced progenitor cell migration toward SMCs through increased SDF-1&agr; production. SDF-1&agr; production by other cell types is regulated by the transcription factor hypoxia-inducible factor (HIF)-1&agr;. We found SMC nuclear HIF-1&agr; expression in PTEN-depleted mice and increased nuclear HIF-1&agr; in PTEN-deficient SMCs. Small interfering RNA–mediated downregulation of HIF-1&agr; reversed SDF-1&agr; induction by PTEN depletion and inhibition of phosphatidylinositol 3-kinase signaling blocked HIF-1&agr; and SDF-1&agr; upregulation induced by PTEN depletion. Our data show that SMC PTEN inactivation establishes an autocrine growth loop and increases progenitor cell recruitment through a HIF-1&agr;–mediated SDF-1&agr;/CXCR4 axis, thus identifying PTEN as a target for the inhibition of pathological vascular remodeling.


Cardiovascular Research | 2010

Inactivation of the tumour suppressor, PTEN, in smooth muscle promotes a pro-inflammatory phenotype and enhances neointima formation

Seth B. Furgeson; Peter Simpson; Insun Park; Vicki VanPutten; Henrick Horita; Christopher D. Kontos; Raphael A. Nemenoff; Mary C.M. Weiser-Evans

AIMS Phosphatase and tensin homolog (PTEN) is implicated as a negative regulator of vascular smooth muscle cell (SMC) proliferation and injury-induced vascular remodelling. We tested if selective depletion of PTEN only in SMC is sufficient to promote SMC phenotypic modulation, cytokine production, and enhanced neointima formation. METHODS AND RESULTS Smooth muscle marker expression and induction of pro-inflammatory cytokines were compared in cultured SMC expressing control or PTEN-specific shRNA. Compared with controls, PTEN-deficient SMC exhibited increased phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signalling and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kappaB) activity, reduced expression of SM markers (SM-alpha-actin and calponin), and increased production of stromal cell-derived factor-1alpha (SDF-1alpha), monocyte chemotactic protein-1 (MCP-1), interleukin-6 (IL-6), and chemokine (C-X-C motif) ligand 1 (KC/CXCL1) under basal conditions. PI3K/Akt or mTOR inhibition reversed repression of SM marker expression, whereas PI3K/Akt or NF-kappaB inhibition blocked cytokine induction mediated by PTEN depletion. Carotid ligation in mice with genetic reduction of PTEN specifically in SMC (SMC-specific PTEN heterozygotes) resulted in enhanced neointima formation, increased SMC hyperplasia, reduced SM-alpha-actin and calponin expression, and increased NF-kappaB and cytokine expression compared with wild-types. Lesion formation in SMC-specific heterozygotes was similar to lesion formation in global PTEN heterozygotes, indicating that inactivation of PTEN exclusively in SMC is sufficient to induce considerable increases in neointima formation. CONCLUSION PTEN activation specifically in SMC is a common upstream regulator of multiple downstream events involved in pathological vascular remodelling, including proliferation, de-differentiation, and production of multiple cytokines.


Seminars in Dialysis | 2008

Review: β-Blockade in Chronic Dialysis Patients

Seth B. Furgeson; Michel Chonchol

Approximately 50% of the mortality in chronic dialysis patients is due to cardiovascular diseases (CVD). Cardiomyopathy, coronary artery disease, and arrhythmia are all common conditions and predispose to sudden death, which accounts for 60% of all cardiac deaths in this population. Despite advances in dialysis therapy, the mortality from CVD remains substantially unchanged, partly due to the lack of evidence‐based strategies for improving the outcome of cardiac diseases in this population. Activation of the sympathetic adrenergic system is well documented in chronic dialysis patients and is likely involved in the pathogenesis of myocardial hypertrophy, coronary artery disease, heart failure, and arrhythmia. Given the proven benefit of β‐blockers in patients with normal kidney function with similar cardiac comorbidities, β‐blockers would seem to be attractive agents to reduce cardiovascular morbidity and mortality in the patient population with advanced chronic kidney disease. However, the value of β‐blockade in patients on chronic dialysis remains unclear. This uncertainty surrounding the efficacy is compounded by the risk of side effects to these patients, such as hypotension, bradycardia, and hyperkalemia. In addition, numerous studies have suggested suboptimal usage of β‐blockers in the dialysis population; this is seen even in high risk patients, such as those with established coronary artery disease. In this review, we will focus on sympathetic nervous system activation in kidney disease and highlight the benefit and risks of β‐blockers usage in the chronic dialysis patient population.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2011

Serum Response Factor Regulates Expression of Phosphatase and Tensin Homolog Through a MicroRNA Network in Vascular Smooth Muscle Cells

Henrick Horita; Peter Simpson; Allison Ostriker; Seth B. Furgeson; Vicki Van Putten; Mary C.M. Weiser-Evans; Raphael A. Nemenoff

Objective—Serum response factor (SRF) is a critical transcription factor in smooth muscle cells (SMCs) controlling differentiation and proliferation. Our previous work demonstrated that depleting SRF in cultured SMCs decreased expression of SMC markers but increased proliferation and inflammatory mediators. A similar phenotype has been observed in SMCs silenced for phosphatase and tensin homolog (PTEN), suggesting that SRF and PTEN may lie on a common pathway. Our goal was to determine the effect of SRF depletion on PTEN levels and define mechanisms mediating this effect. Methods and Results—In SRF-silenced SMCs, PTEN protein levels but not mRNA levels were decreased, suggesting posttranscriptional regulation. Reintroduction of PTEN into SRF-depleted SMCs reversed increases in proliferation and cytokine/chemokine production but had no effect on SMC marker expression. SRF-depleted cells showed decreased levels of microRNA (miR)-143 and increased miR-21, which was sufficient to suppress PTEN. Increased miR-21 expression was dependent on induction of Fos related antigen (FRA)-1, which is a direct target of miR-143. Introducing miR-143 into SRF-depleted SMCs reduced FRA-1 expression and miR-21 levels and restored PTEN expression. Conclusion—SRF regulates PTEN expression in SMCs through a miR network involving miR-143, targeting FRA-1, which regulates miR-21. Cross-talk between SRF and PTEN likely represents a critical axis in phenotypic remodeling of SMCs.


Journal of the American Heart Association | 2013

Selective inactivation of PTEN in smooth muscle cells synergizes with hypoxia to induce severe pulmonary hypertension.

Henrick Horita; Seth B. Furgeson; Allison Ostriker; Kyle A. Olszewski; Timothy M. Sullivan; Leah R. Villegas; Michelle Levine; Jane E. Parr; Carlyne D. Cool; Raphael A. Nemenoff; Mary C.M. Weiser-Evans

Background Pulmonary vascular remodeling in pulmonary hypertension (PH) is characterized by increased vascular smooth muscle cell (SMC) and adventitial fibroblast proliferation, small vessel occlusion, and inflammatory cell accumulation. The underlying molecular mechanisms driving progression remain poorly defined. We have focused on loss of the phosphatase PTEN in SMCs as a major driver of pathological vascular remodeling. Our goal was to define the role of PTEN in human PH and in hypoxia‐induced PH using a mouse model with inducible deletion of PTEN in SMCs. Methods and Results Staining of human biopsies demonstrated enhanced inactive PTEN selectively in the media from hypertensive patients compared to controls. Mice with induced deletion of PTEN in SMCs were exposed to normoxia or hypoxia for up to 4 weeks. Under normoxia, SMC PTEN depletion was sufficient to induce features of PH similar to those observed in wild‐type mice exposed to chronic hypoxia. Under hypoxia, PTEN depletion promoted an irreversible progression of PH characterized by increased pressure, extensive pulmonary vascular remodeling, formation of complex vascular lesions, and increased macrophage accumulation associated with synergistic increases in proinflammatory cytokines and proliferation of both SMCs and nonSMCs. Conclusions Chronic inactivation of PTEN selectively in SMC represents a critical mediator of PH progression, leading to cell autonomous events and increased production of factors correlated to proliferation and recruitment of adventitial and inflammatory cells, resulting in irreversible progression of the disease.


Biomedical Optics Express | 2016

Characterizing fibrosis in UUO mice model using multiparametric analysis of phasor distribution from FLIM images

Suman Ranjit; Alexander S. Dvornikov; Moshe Levi; Seth B. Furgeson; Enrico Gratton

Phasor approach to fluorescence lifetime microscopy is used to study development of fibrosis in the unilateral ureteral obstruction model (UUO) of kidney in mice. Traditional phasor analysis has been modified to create a multiparametric analysis scheme that splits the phasor points in four equidistance segments based on the height of peak of the phasor distribution and calculates six parameters including average phasor positions, the shape of each segment, the angle of the distribution and the number of points in each segment. These parameters are used to create a spectrum of twenty four points specific to the phasor distribution of each sample. Comparisons of spectra from diseased and healthy tissues result in quantitative separation and calculation of statistical parameters including AUC values, positive prediction values and sensitivity. This is a new method in the evolving field of analyzing phasor distribution of FLIM data and provides further insights. Additionally, the progression of fibrosis with time is detected using this multiparametric approach to phasor analysis.


Molecular Pharmacology | 2014

Activation of the Retinoid X Receptor Modulates Angiotensin II-Induced Smooth Muscle Gene Expression and Inflammation in Vascular Smooth Muscle Cells

Allison M.B. Lehman; John R. Montford; Henrick Horita; Allison C. Ostriker; Mary C.M. Weiser-Evans; Raphael A. Nemenoff; Seth B. Furgeson

The retinoid X receptor (RXR) partners with numerous nuclear receptors, such as the peroxisome proliferator activated receptor (PPAR) family, liver X receptors (LXRs), and farnesoid X receptor (FXR). Although each heterodimer can be activated by specific ligands, a subset of these receptors, defined as permissive nuclear receptors, can also be activated by RXR agonists known as rexinoids. Many individual RXR heterodimers have beneficial effects in vascular smooth muscle cells (SMCs). Because rexinoids can potently activate multiple RXR pathways, we hypothesized that treating SMCs with rexinoids would more effectively reverse the pathophysiologic effects of angiotensin II than an individual heterodimer agonist. Cultured rat aortic SMCs were pretreated with either an RXR agonist (bexarotene or 9-cis retinoic acid) or vehicle (dimethylsulfoxide) for 24 hours before stimulation with angiotensin II. Compared with dimethylsulfoxide, bexarotene blocked angiotensin II-induced SM contractile gene induction (calponin and smooth muscle-α-actin) and protein synthesis ([3H]leucine incorporation). Bexarotene also decreased angiotensin II-mediated inflammation, as measured by decreased expression of monocyte chemoattractant protein-1 (MCP-1). Activation of p38 mitogen-activated protein (MAP) kinase but not extracellular signal-related kinase (ERK) or protein kinase B (Akt) was also blunted by bexarotene. We compared bexarotene to five agonists of nuclear receptors (PPARα, PPARγ, PPARδ, LXR, and FXR). Bexarotene had a greater effect on calponin reduction, MCP-1 inhibition, and p38 MAP kinase inhibition than any individual agonist. PPARγ knockout cells demonstrated blunted responses to bexarotene, indicating that PPARγ is necessary for the effects of bexarotene. These data demonstrate that RXR is a potent modulator of angiotensin II-mediated responses in the vasculature, partially through inhibition of p38.


Contributions To Nephrology | 2009

New Treatment Options and Protocols for Peritoneal Dialysis-Related Peritonitis

Seth B. Furgeson; Isaac Teitelbaum

Peritonitis remains a major complication in patients undergoing peritoneal dialysis. The most recent ISPD guidelines for the empiric initial treatment of peritonitis recommend the use of antibiotics that provide coverage against Gram-positive organisms (vancomycin or cefazolin) and Gram-negative organisms (a third-generation cephalosporin or an aminoglycoside). However, there are some situations in which this regimen may not be desirable. Concerns of resistant organisms, changing microbiology, drug toxicity, or difficulties administering therapy may lead a provider to modify the initial regimen. Drug resistant Staphylococcus aureus strains and Enterococcus strains may require administration of newer agents such as linezolid, quinipristin/dalfopristin, or daptomycin. Many centers have reported that, over time, the microbiology at those institutions has been changing. Some centers have reported a significant decrease in gram positive organisms and increase in extended spectrum beta-lactamase (ESBL) organisms. It is important for each center to examine its microbiology to document such trends. Although the currently recommended therapies have low toxicities, it is possible that concerns for untoward side effects in an individual patient may dictate changing the regimen. Finally, there is evidence from many prospective studies that monotherapy with different agents (oral quinolones or cefepime) is efficacious; if ease of therapy is a consideration, these may also be appropriate agents.


Prostaglandins & Other Lipid Mediators | 2016

Cytosolic phospholipase A2α increases proliferation and de-differentiation of human renal tubular epithelial cells.

John R. Montford; Allison Lehman; Micah Scobey; Mary C.M. Weiser-Evans; Raphael A. Nemenoff; Seth B. Furgeson

The group IVA calcium-dependent cytosolic phospholipase A2 (cPLA2α) enzyme controls the release of arachidonic acid from membrane bound phospholipids and is the rate-limiting step in production of eicosanoids. A variety of different kidney injuries activate cPLA2α, therefore we hypothesized that cPLA2α activity would regulate pathologic processes in HK-2 cells, a human renal tubular epithelial cell line, by regulating cell phenotype and proliferation. In two lentiviral cPLA2α-silenced knockdowns, we observed decreased proliferation and increased apoptosis compared to control HK-2 cells. cPLA2α-silenced cells also demonstrated an altered morphology, had increased expression E-cadherin, and decreased expression of Ncadherin. Increased levels of E-cadherin were associated with increased promoter activity and decreased levels of SNAIL1, SNAIL2, and ZEB1, transcriptional repressors of E-cadherin expression. Addition of exogenous arachidonic acid, but not PGE2, reversed the phenotypic changes in cPLA2α-silenced cells. These data suggest that cPLA2α may play a key role in renal repair after injury through a PGE2-independent mechanism.

Collaboration


Dive into the Seth B. Furgeson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Henrick Horita

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Enrico Gratton

University of California

View shared research outputs
Top Co-Authors

Avatar

Isaac Teitelbaum

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Michel Chonchol

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Moshe Levi

University of Colorado Denver

View shared research outputs
Researchain Logo
Decentralizing Knowledge