Network


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

Hotspot


Dive into the research topics where Paul F. Erickson is active.

Publication


Featured researches published by Paul F. Erickson.


Cancer Research | 2008

MicroRNA-137 Targets Microphthalmia-Associated Transcription Factor in Melanoma Cell Lines

Lynne T. Bemis; Robert W. Chen; Carol M. Amato; Elizabeth H. Classen; Steven E. Robinson; David Coffey; Paul F. Erickson; Yiqun G. Shellman; William A. Robinson

Micropthalmia-associated transcription factor (MITF) is the master regulator of melanocyte development, survival, and function. Frequent alteration in the expression of MITF is detected in melanoma, but the mechanism(s) underlying the alteration in expression have not been completely determined. In these studies, we have identified microRNA-137 (miR-137) as a regulator of MITF expression. The genomic locus of miR-137 at chromosome 1p22 places it in a region of the human genome previously determined to harbor an allele for melanoma susceptibility. Here, we show that expression of mature miR-137 in melanoma cell lines down-regulates MITF expression. Further, we have identified a 15-bp variable nucleotide tandem repeat located just 5 to the pre-miR-137 sequence, which alters the processing and function of miR-137 in melanoma cell lines.


Proceedings of the National Academy of Sciences of the United States of America | 2000

Altered HOX and WNT7A expression in human lung cancer

Roser Calvo; James West; Wilbur A. Franklin; Paul F. Erickson; Lynne T. Bemis; Efang Li; Barbara Helfrich; Paul A. Bunn; Joëlle Roche; Elisabeth Brambilla; Rafael Rosell; Robert M. Gemmill; Harry A. Drabkin

HOX genes encode transcription factors that control patterning and cell fates. Alterations in HOX expression have been clearly implicated in leukemia, but their role in most other malignant diseases remains unknown. By using degenerate reverse transcription-PCR and subsequent real-time quantitative assays, we examined HOX expression in lung cancer cell lines, direct tumor-control pairs, and bronchial epithelial cultures. As in leukemia, genes of the HOX9 paralogous group and HOXA10 were frequently overexpressed. For HOXB9, we confirmed that elevated RNA was associated with protein overexpression. In some cases, marked HOX overexpression was associated with elevated FGF10 and FGF17. During development, the WNT pathway affects cell fate, polarity, and proliferation, and WNT7a has been implicated in the maintenance of HOX expression. In contrast to normal lung and mortal short-term bronchial epithelial cultures, WNT7a was frequently reduced or absent in lung cancers. In immortalized bronchial epithelial cells, WNT7a was lost concomitantly with HOXA1, and a statistically significant correlation between the expression of both genes was observed in lung cancer cell lines. Furthermore, we identified a homozygous deletion of beta-catenin in the mesothelioma, NCI-H28, associated with reduced WNT7a and the lowest overall cell line expression of HOXA1, HOXA7, HOXA9, and HOXA10, whereas HOXB9 levels were unaffected. Of note, both WNT7a and beta-catenin are encoded on chromosome 3p, which undergoes frequent loss of heterozygosity in these tumors. Our results suggest that alterations in regulatory circuits involving HOX, WNT, and possibly fibroblast growth factor pathways occur frequently in lung cancer.


Proceedings of the National Academy of Sciences of the United States of America | 2003

WNT7a induces E-cadherin in lung cancer cells.

Tatsuo Ohira; Robert M. Gemmill; Kevin Ferguson; Sophie Kusy; Joëlle Roche; Elisabeth Brambilla; Chan Zeng; Anna E. Barón; Lynne T. Bemis; Paul F. Erickson; Elizabeth L. Wilder; Anil K. Rustgi; Jan Kitajewski; Edward Gabrielson; Roy M. Bremnes; Wilbur A. Franklin; Harry A. Drabkin

E-cadherin loss in cancer is associated with de-differentiation, invasion, and metastasis. Drosophila DE-cadherin is regulated by Wnt/β-catenin signaling, although this has not been demonstrated in mammalian cells. We previously reported that expression of WNT7a, encoded on 3p25, was frequently downregulated in lung cancer, and that loss of E-cadherin or β-catenin was a poor prognostic feature. Here we show that WNT7a both activates E-cadherin expression via a β-catenin specific mechanism in lung cancer cells and is involved in a positive feedback loop. Li+, a GSK3β inhibitor, led to E-cadherin induction in an inositol-independent manner. Similarly, exposure to mWNT7a specifically induced free β-catenin and E-cadherin. Among known transcriptional suppressors of E-cadherin, ZEB1 was uniquely correlated with E-cadherin loss in lung cancer cell lines, and its inhibition by RNA interference resulted in E-cadherin induction. Pharmacologic reversal of E-cadherin and WNT7a losses was achieved with Li+, histone deacetylase inhibition, or in some cases only with combined inhibitors. Our findings provide support that E-cadherin induction by WNT/β-catenin signaling is an evolutionarily conserved pathway operative in lung cancer cells, and that loss of WNT7a expression may be important in lung cancer development or progression by its effects on E-cadherin.


Journal of Biological Chemistry | 2006

Depletion of cAMP-response Element-binding Protein/ATF1 Inhibits Adipogenic Conversion of 3T3-L1 Cells Ectopically Expressing CCAAT/Enhancer-binding Protein (C/EBP) α, C/EBP β, or PPARγ2

Keith E. Fox; Dana M. Fankell; Paul F. Erickson; Susan M. Majka; Joseph T. Crossno; Dwight J. Klemm

The differentiation of preadipocytes to adipocytes is orchestrated by the expression of the “master adipogenic regulators,” CCAAT/enhancer-binding protein (C/EBP) β, peroxisome proliferator-activated receptor γ (PPARγ), and C/EBP α. In addition, activation of the cAMP-response element-binding protein (CREB) is necessary and sufficient to promote adipogenic conversion and prevent apoptosis of mature adipocytes. In this report we used small interfering RNAto deplete CREB and the closely related factor ATF1 to explore the ability of the master adipogenic regulators to promote adipogenesis in the absence of CREB and probe the function of CREB in late stages of adipogenesis. Loss of CREB/ATF1 blocked adipogenic conversion of 3T3-L1 cells in culture or 3T3-F442A cells implanted into athymic mice. Loss of CREB/ATF1 prevented the expression of PPARγ, C/EBP α, and adiponectin and inhibited the loss of Pref-1. Loss of CREB/ATF1 inhibited adipogenic conversion even in cells ectopically expressing C/EBP α, C/EBP β, or PPARγ2 individually. CREB/ATF1 depletion did not attenuate lipid accumulation in cells expressing both PPARγ2 and C/EBP α, but adiponectin expression was severely diminished. Conversely ectopic expression of constitutively active CREB overcame the blockade of adipogenesis due to depletion of C/EBP β but not due to loss of PPARγ2 or C/EBP α. Depletion of CREB/ATF1 did not suppress the expression of C/EBP β as we had previously observed using dominant negative forms of CREB. Finally results are presented showing that CREB promotes PPARγ2 gene transcription. The results indicate that CREB and ATF1 play a central role in adipogenesis because expression of individual master adipogenic regulators is unable to compensate for their loss. The data also indicate that CREB not only functions during the initiation of adipogenic conversion but also at later stages.


Oncogene | 2002

The TRC8 hereditary kidney cancer gene suppresses growth and functions with VHL in a common pathway

Robert M. Gemmill; Lynne T. Bemis; Jason P. Lee; M Ali Sozen; Anna E. Barón; Chan Zeng; Paul F. Erickson; Joan E. Hooper; Harry A. Drabkin

VHL is part of an SCF related E3-ubiquitin ligase complex with ‘gatekeeper’ function in renal carcinoma. However, no mutations have been identified in VHL interacting proteins in wild type VHL tumors. We previously reported that the TRC8 gene was interrupted by a t(3;8) translocation in a family with hereditary renal and non-medullary thyroid cancer. TRC8 encodes a multi-membrane spanning protein containing a RING-H2 finger with in vitro ubiquitin ligase activity. We isolated the Drosophila homologue, DTrc8, and studied its function by genetic manipulations and a yeast 2-hybrid screen. Human and Drosophila TRC8 proteins localize to the endoplasmic reticulum. Loss of either DTrc8 or DVhl resulted in an identical ventral midline defect. Direct interaction between DTrc8 and DVhl was confirmed by GST-pulldown and co-immunoprecipitation experiments. CSN-5/JAB1 is a component of the COP9 signalosome, recently shown to regulate SCF function. We found that DTrc8 physically interacts with CSN-5 and that human JAB1 localization is dependent on VHL mutant status. Lastly, overexpression of DTrc8 inhibited growth consistent with its presumed role as a tumor suppressor gene. Thus, VHL, TRC8, and JAB1 appear to be linked both physically and functionally and all three may participate in the development of kidney cancer.


Molecular and Cellular Biology | 2006

Platelet-Derived Growth Factor BB Induces Nuclear Export and Proteasomal Degradation of CREB via Phosphatidylinositol 3-Kinase/Akt Signaling in Pulmonary Artery Smooth Muscle Cells

Chrystelle V. Garat; Dana M. Fankell; Paul F. Erickson; Jane E.B. Reusch; Natalie N. Bauer; Ivan F. McMurtry; Dwight J. Klemm

ABSTRACT Cyclic AMP response element binding protein (CREB) content is diminished in smooth muscle cells (SMCs) in remodeled pulmonary arteries from animals with pulmonary hypertension and in the SMC layers of atherogenic systemic arteries and cardiomyocytes from hypertensive individuals. Loss of CREB can be induced in cultured SMCs by chronic exposure to hypoxia or platelet-derived growth factor BB (PDGF-BB). Here we investigated the signaling pathways and mechanisms by which PDGF elicits depletion of SMC CREB. Chronic PDGF treatment increased CREB ubiquitination in SMCs, while treatment of SMCs with the proteasome inhibitor lactacystin prevented decreases in CREB content. The nuclear export inhibitor leptomycin B also prevented depletion of SMC CREB alone or in combination with lactacystin. Subsequent studies showed that PDGF activated extracellular signal-regulated kinase, Jun N-terminal protein kinase, and phosphatidylinositol 3 (PI3)-kinase pathways in SMCs. Inhibition of these pathways blocked SMC proliferation in response to PDGF, but only inhibition of PI3-kinase or its effector, Akt, blocked PDGF-induced CREB loss. Finally, chimeric proteins containing enhanced cyan fluorescent protein linked to wild-type CREB or CREB molecules with mutations in several recognized phosphorylation sites were introduced into SMCs. PDGF treatment reduced the levels of each of these chimeric proteins except for one containing mutations in adjacent serine residues (serines 103 and 107), suggesting that CREB loss was dependent on CREB phosphorylation at these sites. We conclude that PDGF stimulates nuclear export and proteasomal degradation of CREB in SMCs via PI3-kinase/Akt signaling. These results indicate that in addition to direct phosphorylation, proteolysis and intracellular localization are key mechanisms regulating CREB content and activity in SMCs.


Journal of Biological Chemistry | 2008

Regulation of Cyclin D1 and Wnt10b Gene Expression by cAMP-responsive Element-binding Protein during Early Adipogenesis Involves Differential Promoter Methylation

Keith E. Fox; Lillester A. Colton; Paul F. Erickson; Jacob E. Friedman; Hyuk C. Cha; Pernille Keller; Ormond A. MacDougald; Dwight J. Klemm

Cyclin D1 expression is elevated and Wnt10b is repressed by cAMP during the first few hours of adipogenesis. cAMP-responsive element-binding protein (CREB) is a primary target for cAMP signaling, and we have shown that activation of CREB promotes adipogenesis and adipocyte survival. Here we tested the impact of CREB on expression of cyclin D1 and wingless-related mouse mammary tumor virus integration site 10b (Wnt10b) in 3T3-L1 cells. Forced depletion of CREB blocked Bt2cAMP-stimulated cyclin D1 expression and basal Wnt10b gene expression. Two CREB-binding sites were identified in the Wnt10b promoter region. Ablation of either site partially blocked promoter activity, while mutation of both sites completely suppressed promoter activity. These results suggest that CREB activates transcription from both the cyclin D1 and Wnt10b gene promoters. What accounts for the differential regulation of cyclin D1 and Wnt10b genes by cAMP? Chromatin immunoprecipitation revealed CREB bound to the Wnt10b promoter in untreated preadipocytes but not following treatment with Bt2cAMP. CREB binding to the cyclin D1 promoter was detected in untreated cells and post-Bt2cAMP. Differences between CREB binding to the two genes correlated with increasing methylation of the Wnt10b promoter following Bt2cAMP treatment, whereas no methylation of the cyclin D1 promoter was observed. Treatment of cells with the methylase inhibitor 5-azacytidine restored CREB binding to the Wnt10b gene promoter and prevented the inhibition of Wnt10b RNA expression by Bt2cAMP. We conclude that cAMP stimulates phosphorylation and binding of CREB to the cyclin D1 gene promoter. Simultaneously, hypermethylation of the Wnt10b gene promoter suppresses binding of CREB, allowing adipogenesis to proceed.


Journal of Neurochemistry | 1980

Axonal transport of the Ca2+-dependent protein modulator of 3':5'-cyclic-AMP phosphodiesterase in the rabbit visual system.

Paul F. Erickson; Kenneth B. Seamon; Blake W. Moore; Robert S. Lasher; Lee N. Minier

Abstract: Water‐soluble proteins were extracted from individual retinas, optic nerves, combined optic tracts and lateral geniculate bodies, and superior colliculi of rabbits at 1, 3, and 18 days after injection of [3H]leucine into the right eye. The Ca2+‐dependent protein modulator of 3′:5′‐cyclic‐AMP phos‐phodiesterase (calmodulin) was isolated from these samples by a two‐step polyacrylamide gel electrophoresis procedure. An analysis of the radioactivity incorporated into the total soluble proteins and the calmodulin revealed that most of the calmodulin was axonally transported at a slow rate (2–4 mm/day) and represented about 0.45% of the total transported soluble protein.


The FASEB Journal | 2016

De novo generation of adipocytes from circulating progenitor cells in mouse and human adipose tissue

Kathleen M. Gavin; Jonathan A. Gutman; Wendy M. Kohrt; Qi Wei; Karen L. Shea; Heidi L. Miller; Timothy M. Sullivan; Paul F. Erickson; Karen M. Helm; Alistaire S. Acosta; Christine R. Childs; Evelyn Musselwhite; Marileila Varella-Garcia; Kimberly Kelly; Susan M. Majka; Dwight J. Klemm

White adipocytes in adults are typically derived from tissue resident mesenchymal progenitors. The recent identification of de novo production of adipocytes from bone marrow progenitor‐derived cells in mice challenges this paradigm and indicates an alternative lineage specification that adipocytes exist. We hypothesized that alternative lineage specification of white adipocytes is also present in human adipose tissue. Bone marrow from transgenic mice in which luciferase expression is governed by the adipocyte‐restricted adiponectin gene promoter was adoptively transferred to wild‐type recipient mice. Light emission was quantitated in recipients by in vivo imaging and direct enzyme assay. Adipocytes were also obtained from human recipients of hematopoietic stem cell transplantation. DNA was isolated, and microsatellite polymorphisms were exploited to quantify donor/recipient chimerism. Luciferase emission was detected from major fat depots of transplanted mice. No light emission was observed from intestines, liver, or lungs. Up to 35% of adipocytes in humans were generated from donor marrow cells in the absence of cell fusion. Nontransplanted mice and stromal‐vascular fraction samples were used as negative and positive controls for the mouse and human experiments, respectively. This study provides evidence for a nontissue resident origin of an adipocyte subpopulation in both mice and humans.—Gavin, K. M., Gutman, J. A., Kohrt, W. M., Wei, Q., Shea, K. L., Miller, H. L., Sullivan, T. M., Erickson, P. F., Helm, K. M., Acosta, A. S., Childs, C. R., Musselwhite, E., Varella‐Garcia, M., Kelly, K., Majka, S. M., Klemm, D. J., De novo generation of adipocytes from circulating progenitor cells in mouse and human adipose tissue. FASEB J. 30, 1096–1108 (2016). www.fasebj.org


Proceedings of the National Academy of Sciences of the United States of America | 1993

The 3;21 translocation in myelodysplasia results in a fusion transcript between the AML1 gene and the gene for EAP, a highly conserved protein associated with the Epstein-Barr virus small RNA EBER 1

Giuseppina Nucifora; Catherine R. Begy; Paul F. Erickson; Harry A. Drabkin; Janet D. Rowley

Collaboration


Dive into the Paul F. Erickson's collaboration.

Top Co-Authors

Avatar

Harry A. Drabkin

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Robert S. Lasher

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Dwight J. Klemm

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert M. Gemmill

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna E. Barón

Colorado School of Public Health

View shared research outputs
Top Co-Authors

Avatar

Chan Zeng

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Keith E. Fox

University of Colorado Denver

View shared research outputs
Researchain Logo
Decentralizing Knowledge