Network


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

Hotspot


Dive into the research topics where Erich J. Kushner is active.

Publication


Featured researches published by Erich J. Kushner.


International Journal of Obesity | 2009

Endothelial progenitor cell number and colony-forming capacity in overweight and obese adults

Owen J. MacEneaney; Erich J. Kushner; G P Van Guilder; Jared J. Greiner; Brian L. Stauffer; Christopher A. DeSouza

Objective:To investigate whether adiposity influences endothelial progenitor cell (EPC) number and colony-forming capacity.Design:Cross-sectional study of normal weight, overweight and obese adult humans.Participants:Sixty-seven sedentary adults (aged 45–65 years): 25 normal weight (body mass index (BMI) ⩽25 kg/m2; 12 males/13 females); 18 overweight (BMI=25–29.9 kg/m2; 12 males/6 females); and 24 obese (BMI ⩾30 kg/m2; 18 males/6 females). All participants were non-smokers and free of overt cardiometabolic disease.Measurements:Peripheral blood samples were collected and circulating EPC number was assessed by flow cytometry. Putative EPCs were defined as CD45−/CD34+/VEGFR-2+/CD133+ or CD45−/CD34+ cells. EPC colony-forming capacity was measured in vitro using a colony-forming unit (CFU) assay.Results:Number of circulating putative EPCs (either CD45−/CD34+/VEGFR-2+/CD133+ or CD45−/CD34+ cells) was lower (P<0.05) in obese (0.0007±0.0001%; 0.050±0.006%) compared with overweight (0.0016±0.0004%; 0.089±0.019%) and normal weight (0.0015±0.0003%; 0.082±0.008%) adults. There were no differences in EPC number between the overweight and normal weight groups. EPC colony formation was significantly less in the obese (6±1) and overweight (4±1) compared with normal weight (9±2) adults.Conclusion:These results indicate that: (1) the number of circulating EPCs is lower in obese compared with overweight and normal weight adults; and (2) EPC colony-forming capacity is blunted in overweight and obese adults compared with normal weight adults. Impairments in EPC number and function may contribute to adiposity-related cardiovascular risk.


Clinical Chemistry and Laboratory Medicine | 2009

Aging and Endothelial Progenitor Cell Telomere Length in Healthy Men

Erich J. Kushner; Gary P. Van Guilder; Owen J. MacEneaney; Jennifer N. Cech; Brian L. Stauffer; Christopher A. DeSouza

Abstract Background: Telomere length declines with age in mature endothelial cells and is thought to contribute to endothelial dysfunction and atherogenesis. Bone marrow-derived circulating endothelial progenitor cells (EPCs) are critical to vascular health as they contribute to both reendothelialization and neovascularization. We tested the hypothesis that EPC telomere length decreases with age in healthy adult humans. Methods: Peripheral blood samples were collected from 40 healthy, non-obese, sedentary men: 12 young (age 21–34 years), 12 middle-aged (43–55 years) and 16 older (57–68 years). Putative EPCs were isolated from peripheral blood mononuclear cells and telomere length was determined using genomic DNA preparation and Southern hybridization techniques. Results: EPC telomere length (base pairs) was ∼20% (p=0.01) lower in the older (8492+523 bp) compared to the middle-aged (10,565+572 bp) and young (10,205+501 bp) men. Of note, there was no difference in EPC telomere length between the middle-aged and young men. Conclusions: These results demonstrate that EPC telomere length declines with age in healthy, sedentary men. Interestingly, telomere length was well preserved in the middle-aged compared to young men, suggesting that EPC telomere shortening occurs after the age of 55 years. Clin Chem Lab Med 2009;47:47–50.


Current Opinion in Hematology | 2013

Building blood vessels in development and disease.

Erich J. Kushner; Victoria L. Bautch

Purpose of reviewThis review will examine developmental angiogenesis and tumor-related changes to endothelial cells. Recent findingsProcesses that govern developmental angiogenesis become dysfunctional in the tumor environment, leading to abnormal tumor endothelial cells and blood vessels. Recent findings suggest that tumor endothelial cells are permanently modified compared with normal counterparts. SummaryCoordination of numerous intracellular and extracellular programs promotes the formation of new blood vessels that are necessary for both development and certain diseases. Developmental angiogenesis uses canonical signaling modalities to effectively assemble endothelial cells into predictable vessel structures, and disruption of critical signaling factors has dramatic effects on blood vessel development. Solid tumors co-opt developmental cues to promote formation of tumor vessels that sustain their growth, but these angiogenic signals are not well regulated and produce endothelial cell dysfunction. Aberrant growth factor signaling contributes to phenotypic changes and acquired irreversible intracellular signaling, cytoskeletal and genetic modifications in endothelial cells of tumor vessels. Permanently altered tumor endothelial cells may represent a significant population.


Journal of Vascular Research | 2011

Aging Is Associated with a Proapoptotic Endothelial Progenitor Cell Phenotype

Erich J. Kushner; Owen J. MacEneaney; Brian R. Weil; Jared J. Greiner; Brian L. Stauffer; Christopher A. DeSouza

The aim of this study was to determine if aging is associated with enhanced endothelial progenitor cell (EPC) sensitivity to apoptosis. Cells with phenotypic EPC characteristics were isolated from healthy, nonobese young (age 25 ± 1 years) and older (61 ± 1 years) men. Intracellular active caspase-3 concentrations in response to staurosporine stimulation were approximately 35% higher (p < 0.05) in EPCs from older (3.15 ± 0.29 pg/ml) compared with young (2.33 ± 0.24 pg/ml) men. Protein expression of Akt, p70 S6-kinase and Bcl-2 was markedly lower (approx. 35, 75 and 60%, respectively, all p < 0.05) in EPCs from older compared with young men, whereas there were no age-related differences in either 14-3-3Ε or Bax expression. Additionally, EPC telomerase activity was 57% lower (p < 0.05) in older (0.18 ± 0.11 AU) versus young (0.43 ± 0.11 AU) men. These results indicate that aging is associated with a proapoptotic EPC phenotype characterized by decreased expression of key antiapoptotic proteins associated with the PI-3-kinase signaling pathway and reduced telomerase activity. These age-related changes likely contribute, in part, to the diminished ability of EPCs to resist an apoptotic stimulus in older men. Increased susceptibility to apoptosis may contribute to the numerical and functional impairments observed in EPCs with aging.


Developmental Cell | 2014

Decoy Receptor CXCR7 Modulates Adrenomedullin-Mediated Cardiac and Lymphatic Vascular Development

Klara R. Klein; Natalie O. Karpinich; Scott T. Espenschied; Helen H. Willcockson; William P. Dunworth; Samantha L. Hoopes; Erich J. Kushner; Victoria L. Bautch; Kathleen M. Caron

Atypical 7-transmembrane receptors, often called decoy receptors, act promiscuously as molecular sinks to regulate ligand bioavailability and consequently temper the signaling of canonical G protein-coupled receptor (GPCR) pathways. Loss of mammalian CXCR7, the most recently described decoy receptor, results in postnatal lethality due to aberrant cardiac development and myocyte hyperplasia. Here, we provide the molecular underpinning for this proliferative phenotype by demonstrating that the dosage and signaling of adrenomedullin (Adm, gene; AM, protein)-a mitogenic peptide hormone required for normal cardiovascular development-is tightly controlled by CXCR7. To this end, Cxcr7(-/-) mice exhibit gain-of-function cardiac and lymphatic vascular phenotypes that can be reversed upon genetic depletion of adrenomedullin ligand. In addition to identifying a biological ligand accountable for the phenotypes of Cxcr7(-/-) mice, these results reveal a previously underappreciated role for decoy receptors as molecular rheostats in controlling the timing and extent of GPCR-mediated cardiac and vascular development.


Obesity | 2010

Endothelial Progenitor Cell Function, Apoptosis, and Telomere Length in Overweight/Obese Humans

Owen J. MacEneaney; Erich J. Kushner; Christian M. Westby; Jennifer N. Cech; Jared J. Greiner; Brian L. Stauffer; Christopher A. DeSouza

Excess adiposity is associated with increased cardiovascular morbidity and mortality. Endothelial progenitor cells (EPCs) play an important role in vascular repair. We tested the hypothesis that increased adiposity is associated with EPC dysfunction, characterized by diminished capacity to release angiogenic cytokines, increased apoptotic susceptibility, reduced cell migration, and shorter telomere length. A total of 67 middle‐aged and older adults (42–67 years) were studied: 25 normal weight (normal weight; BMI: 18.5–24.9 kg/m2) and 42 overweight/obese (overweight/obese; BMI: 25.0–34.9 kg/m2). Cells with phenotypic EPC characteristics were isolated from peripheral blood. EPC release of vascular endothelial growth factor (VEGF) and granulocyte colony–stimulating factor (G‐CSF) was determined in the absence and presence of phytohemagglutinin (10 µg/ml). Intracellular active caspase‐3 and cytochrome c concentrations were determined by immunoassay. Migratory activity of EPCs in response to VEGF (2 ng/ml) and stromal cell–derived factor‐1α (SDF‐1α; 10 ng/ml) was determined by Boyden chamber. Telomere length was assessed by Southern hybridization. Phytohemagglutinin‐stimulated release of VEGF (90.6 ± 7.6 vs. 127.2 ± 11.6 pg/ml) and G‐CSF (896.1 ± 77.4 vs. 1,176.3 ± 126.3 pg/ml) was ∼25% lower (P < 0.05) in EPCs from overweight/obese vs. normal weight subjects. Staurosporine induced a ∼30% greater (P < 0.05) increase in active caspase‐3 in EPCs from overweight/obese (2.8 ± 0.2 ng/ml) compared with normal weight (2.2 ± 0.2) subjects. There were no significant differences in EPC migration to either VEGF or SDF‐1α. Telomere length did not differ between groups. These results indicate that increased adiposity adversely affects the ability of EPCs to release proangiogenic cytokines and resist apoptosis, potentially compromising their reparative potential.


Journal of Cell Biology | 2014

Excess centrosomes disrupt endothelial cell migration via centrosome scattering

Erich J. Kushner; Luke S. Ferro; Jie Yu Liu; Jessica R. Durrant; Stephen L. Rogers; Andrew C. Dudley; Victoria L. Bautch

Centrosome–microtubule interactions during interphase are important for centrosome clustering and cell polarity.


Blood Cells Molecules and Diseases | 2010

CD31+ T cells represent a functionally distinct vascular T cell phenotype

Erich J. Kushner; Owen J. MacEneaney; Richard G. Morgan; Alexander M. Van Engelenburg; Gary P. Van Guilder; Christopher A. DeSouza

In contrast to CD3(+)/CD31(-) cells, CD3(+)/CD31(+) cells aid in endothelial repair and revascularization. There are limited data regarding the functional differences between circulating CD3(+)/CD31(+) and CD3(+)/CD31(-) cells that may contribute to their divergent cardiovascular effects. The aim of the present study was to characterize functional differences between CD3(+)/CD31(+) and CD3(+)/CD31(-) cells. To address this aim, migratory capacity, proangiogenic cytokine release and apoptotic susceptibility of CD3(+)/CD31(+) and CD3(+)/CD31(-) cells were determined. Human CD3(+)/CD31(+) and CD3(+)/CD31(-)cells from peripheral blood were isolated using magnetic-activated cell sorting. CD3(+)/CD31(+) cells demonstrated significantly higher ( approximately 60%) migratory capacity to the chemokines SDF-1alpha (655+/-99 vs. 273+/-54 AU) and VEGF (618+/-99 vs. 259+/-57 AU) vs. CD3(+)/CD31(-) cells. Release of angiogenic cytokines G-CSF, interleukin-8 and matrix metallopeptidase-9 were all approximately 100% higher (P<0.05) in CD3(+)/CD31(+) than CD3(+)/CD31(-) cells. CD3(+)/CD31(+) cells exhibited significantly higher intracellular concentrations of active caspase-3 (2.61+/-0.60 vs. 0.34+/-0.09 ng/mL) and cytochrome-c (21.8+/-1.4 vs. 13.7+/-1.0 ng/mL). In summary, CD3(+)/CD31(+) cells have greater migratory and angiogenic cytokine release capacity, but are more susceptible to apoptosis compared with CD3(+)/CD31(-) cells. Enhanced migratory capacity and angiogenic cytokine release may contribute to the vasculogenic properties of this unique T cell subpopulation.


Journal of Human Hypertension | 2011

Prehypertension and Endothelial Progenitor Cell Function.

Owen J. MacEneaney; Christopher A. DeSouza; Brian R. Weil; Erich J. Kushner; G P Van Guilder; Michael L. Mestek; Jared J. Greiner; Brian L. Stauffer

Prehypertension is associated with significant damage to the coronary vasculature and increased rates of adverse cardiovascular events. Circulating endothelial progenitor cells (EPCs) are critical to vascular repair and the formation of new blood vessels. We tested the hypothesis that prehypertension is associated with EPC dysfunction. Peripheral blood samples were collected from 83 middle-aged and older adults (51 male and 32 female): 40 normotensive subjects (age 53±2 years; BP 111/74±1/1 mm Hg) and 43 prehypertensive subjects (age 54±2 years; 128/77±1/1 mm Hg). EPCs were isolated from peripheral blood, and EPC colony-forming capacity (colony-forming unit (CFU) assay), migratory activity (Boyden chamber) and apoptotic susceptibility (active caspase-3 concentrations) were determined. There were no significant differences in the number of EPC CFUs (10±2 vs 9±1), EPC migration (1165±82 vs 1120±84 fluorescent units) or active intracellular caspase-3 concentrations (2.7±0.3 vs 2.3±0.2 ng ml−1) between the normotensive and prehypertensive groups. When groups were stratified into low prehypertension (n=27; systolic blood pressure: 120–129 mm Hg) and high prehypertension (n=16; 130–139 mm Hg), it was found that EPCs from the high prehypertensive group produced fewer (∼65%, P<0.05) CFUs compared with the low prehypertensive (4±1 vs 12±2) and normotensive adults. In conclusion, EPC colony-forming capacity is impaired only in prehypertensive adults with systolic BP greater than 130 mm Hg. Prehypertension is not associated with migratory dysfunction or enhanced apoptosis of EPCs.


Nature Communications | 2016

Notch regulates BMP responsiveness and lateral branching in vessel networks via SMAD6

Kevin P. Mouillesseaux; David S. Wiley; Lauren M. Saunders; Lyndsay A. Wylie; Erich J. Kushner; Diana C. Chong; Kathryn M. Citrin; Andrew T. Barber; Youngsook Park; Jun Dae Kim; Leigh Ann Samsa; Jongmin Kim; Jiandong Liu; Suk-Won Jin; Victoria L. Bautch

Functional blood vessel growth depends on generation of distinct but coordinated responses from endothelial cells. Bone morphogenetic proteins (BMP), part of the TGFβ superfamily, bind receptors to induce phosphorylation and nuclear translocation of SMAD transcription factors (R-SMAD1/5/8) and regulate vessel growth. However, SMAD1/5/8 signalling results in both pro- and anti-angiogenic outputs, highlighting a poor understanding of the complexities of BMP signalling in the vasculature. Here we show that BMP6 and BMP2 ligands are pro-angiogenic in vitro and in vivo, and that lateral vessel branching requires threshold levels of R-SMAD phosphorylation. Endothelial cell responsiveness to these pro-angiogenic BMP ligands is regulated by Notch status and Notch sets responsiveness by regulating a cell-intrinsic BMP inhibitor, SMAD6, which affects BMP responses upstream of target gene expression. Thus, we reveal a paradigm for Notch-dependent regulation of angiogenesis: Notch regulates SMAD6 expression to affect BMP responsiveness of endothelial cells and new vessel branch formation.

Collaboration


Dive into the Erich J. Kushner's collaboration.

Top Co-Authors

Avatar

Christopher A. DeSouza

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Owen J. MacEneaney

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Brian L. Stauffer

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Jared J. Greiner

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Victoria L. Bautch

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gary P. Van Guilder

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Jennifer N. Cech

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Christian M. Westby

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Kyle J. Diehl

University of Colorado Boulder

View shared research outputs
Researchain Logo
Decentralizing Knowledge