Network


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

Hotspot


Dive into the research topics where Russell Ericksen is active.

Publication


Featured researches published by Russell Ericksen.


Gastroenterology | 2013

Mice That Express Human Interleukin-8 Have Increased Mobilization of Immature Myeloid Cells, Which Exacerbates Inflammation and Accelerates Colon Carcinogenesis

Samuel Asfaha; Alexander Dubeykovskiy; Hiroyuki Tomita; Xiangdong Yang; Sarah Stokes; Wataru Shibata; Richard A. Friedman; Hiroshi Ariyama; Zinaida A. Dubeykovskaya; Sureshkumar Muthupalani; Russell Ericksen; Harold Frucht; James G. Fox; Timothy C. Wang

BACKGROUND & AIMS Interleukin (IL)-8 has an important role in initiating inflammation in humans, attracting immune cells such as neutrophils through their receptors CXCR1 and CXCR2. IL-8 has been proposed to contribute to chronic inflammation and cancer. However, mice do not have the IL-8 gene, so human cancer cell lines and xenograft studies have been used to study the role of IL-8 in colon and gastric carcinogenesis. We generated mice that carry a bacterial artificial chromosome that encompasses the entire human IL-8 gene, including its regulatory elements (IL-8Tg mice). METHODS We studied the effects of IL-8 expression in APCmin(+/-) mice and IL-8Tg mice given azoxymethane and dextran sodium sulfate (DSS). We also examined the effects of IL-8 expression in gastric cancer in INS-GAS mice that overexpress gastrin and IL-8Tg mice infected with Helicobacter felis. RESULTS In IL-8Tg mice, expression of human IL-8 was controlled by its own regulatory elements, with virtually no messenger RNA or protein detectable under basal conditions. IL-8 was strongly up-regulated on systemic or local inflammatory stimulation, increasing mobilization of immature CD11b(+)Gr-1(+) myeloid cells (IMCs) with thioglycolate-induced peritonitis, DSS-induced colitis, and H. felis-induced gastritis. IL-8 was increased in colorectal tumors from patients and IL-8Tg mice compared with nontumor tissues. IL-8Tg mice developed more tumors than wild-type mice following administration of azoxymethane and DSS. Expression of IL-8 increased tumorigenesis in APCmin(+/-) mice compared with APCmin(+/-) mice that lack IL-8; this was associated with increased numbers of IMCs and angiogenesis in the tumors. CONCLUSIONS IL-8 contributes to gastrointestinal carcinogenesis by mobilizing IMCs and might be a therapeutic target for gastrointestinal cancers.


Cell Stem Cell | 2015

Krt19+/Lgr5− Cells Are Radioresistant Cancer-Initiating Stem Cells in the Colon and Intestine

Samuel Asfaha; Yoku Hayakawa; Ashlesha Muley; Sarah Stokes; Trevor A. Graham; Russell Ericksen; Christoph B. Westphalen; Johannes von Burstin; Teresa L. Mastracci; Daniel L. Worthley; Chandhan Guha; Michael Quante; Anil K. Rustgi; Timothy C. Wang

Epithelium of the colon and intestine are renewed every 3 days. In the intestine there are at least two principal stem cell pools. The first contains rapid cycling crypt-based columnar (CBC) Lgr5(+) cells, and the second is composed of slower cycling Bmi1-expressing cells at the +4 position above the crypt base. In the colon, however, the identification of Lgr5(-) stem cell pools has proven more challenging. Here, we demonstrate that the intermediate filament keratin-19 (Krt19) marks long-lived, radiation-resistant cells above the crypt base that generate Lgr5(+) CBCs in the colon and intestine. In colorectal cancer models, Krt19(+) cancer-initiating cells are also radioresistant, while Lgr5(+) stem cells are radiosensitive. Moreover, Lgr5(+) stem cells are dispensable in both the normal and neoplastic colonic epithelium, as ablation of Lgr5(+) stem cells results in their regeneration from Krt19-expressing cells. Thus, Krt19(+) stem cells are a discrete target relevant for cancer therapy.


Cancer Research | 2010

K-ras Mutation Targeted to Gastric Tissue Progenitor Cells Results in Chronic Inflammation, an Altered Microenvironment, and Progression to Intraepithelial Neoplasia

Tomoyuki Okumura; Russell Ericksen; Shigeo Takaishi; Sophie S.W. Wang; Zinaida Dubeykovskiy; Wataru Shibata; Kelly S. Betz; Sureshkuma Muthupalani; Arlin B. Rogers; James G. Fox; Anil K. Rustgi; Timothy C. Wang

Chronic infectious diseases, such as Helicobacter pylori infection, can promote cancer in a large part through induction of chronic inflammation. Oncogenic K-ras mutation in epithelial cells activates inflammatory pathways, which could compensate for a lack of infectious stimulus. Gastric histopathology and putative progenitor markers [doublecortin and calcium/calmodulin-dependent protein kinase-like 1 (Dcamkl1) and keratin 19 (K19)] in K19-K-ras-V12 (K19-kras) transgenic mice were assessed at 3, 6, 12, and 18 months of age, in comparison with Helicobacter felis-infected wild-type littermates. Inflammation was evaluated by reverse transcription-PCR of proinflammatory cytokines, and K19-kras mice were transplanted with green fluorescent protein (GFP)-labeled bone marrow. Both H. felis infection and K-ras mutation induced upregulation of proinflammatory cytokines, expansion of Dcamkl1(+) cells, and progression to oxyntic atrophy, metaplasia, hyperplasia, and high-grade dysplasia. K19-kras transgenic mice uniquely displayed mucous metaplasia as early as 3 months and progressed to high-grade dysplasia and invasive intramucosal carcinoma by 20 months. In bone marrow-transplanted K19-kras mice that progressed to dysplasia, a large proportion of stromal cells were GFP(+) and bone marrow-derived, but only rare GFP(+) epithelial cells were observed. GFP(+) bone marrow-derived cells included leukocytes and CD45(-) stromal cells that expressed vimentin or α smooth muscle actin and were often found surrounding clusters of Dcamkl1(+) cells at the base of gastric glands. In conclusion, the expression of mutant K-ras in K19(+) gastric epithelial cells can induce chronic inflammation and promote the development of dysplasia.


Laboratory Investigation | 2009

Identification of a bone marrow-derived mesenchymal progenitor cell subset that can contribute to the gastric epithelium

Tomoyuki Okumura; Sophie S.W. Wang; Shigeo Takaishi; Shui Ping Tu; Vivian G. Ng; Russell Ericksen; Anil K. Rustgi; Timothy C. Wang

Recent studies with Helicobacter-infected mice have shown that bone marrow-derived cells can repopulate the gastric epithelium and progress to cancer. However, it has not been established which cellular subset can potentially contribute to the epithelium. The aim of this study was to investigate the ability of bone marrow-derived mesenchymal stem cells (MSCs) that express cytokeratin 19 (K19) to contribute to the gastric epithelium. MSCs cultures were established from whole bone marrow and expression of K19 was detected in a minority (1 of 13) of clones by real-time PCR and immunostaining. Transfection of a K19-green fluorescent protein (GFP) vector and isolation of GFP-expressing colonies generated high K19-expressing MSC clones (K19GFPMSC). Incubation of MSCs with gastric tissue extract markedly induced mRNA expression of gastric phenotypic markers and was observed to a greater extent in K19GFPMSCs compared with parental MSCs and mock transfectants. Both K19GFPMSCs and GFP-labeled control MSCs gave rise to gastric epithelial cells after injection into the murine stomach. In addition, after blastocyst injections, K19GFPMSCs gave rise to GFP-positive gastric epithelial cells in all 13 pups, whereas only 3 of 10 offspring showed GFP-positive gastric epithelial cells after injection of GFP-labeled control MSCs. Although K19 expression could not be detected in murine whole bone marrow, H. felis infection increased K19-expressing MSCs in the circulation. Taken together, our results show that bone marrow-derived MSCs can contribute to the gastric epithelium. The K19-positive MSC fraction that is induced by chronic H. felis infection appears to be the important subset in this process.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2011

In vivo analysis of mouse gastrin gene regulation in enhanced GFP-BAC transgenic mice.

Shigeo Takaishi; Wataru Shibata; Hiroyuki Tomita; Guangchun Jin; Xiangdong Yang; Russell Ericksen; Zinaida A. Dubeykovskaya; Samuel Asfaha; Michael Quante; Kelly S. Betz; Arthur Shulkes; Timothy C. Wang

Gastrin is secreted from a subset of neuroendocrine cells residing in the gastric antrum known as G cells, but low levels are also expressed in fetal pancreas and intestine and in many solid malignancies. Although past studies have suggested that antral gastrin is transcriptionally regulated by inflammation, gastric pH, somatostatin, and neoplastic transformation, the transcriptional regulation of gastrin has not previously been demonstrated in vivo. Here, we describe the creation of an enhanced green fluorescent protein reporter (mGAS-EGFP) mouse using a bacterial artificial chromosome that contains the entire mouse gastrin gene. Three founder lines expressed GFP signals in the gastric antrum and the transitional zone to the corpus. In addition, GFP(+) cells could be detected in the fetal pancreatic islets and small intestinal villi, but not in these organs of the adult mice. The administration of acid-suppressive reagents such as proton pump inhibitor omeprazole and gastrin/CCK-2 receptor antagonist YF476 significantly increased GFP signal intensity and GFP(+) cell numbers in the antrum, whereas these parameters were decreased by overnight fasting, octreotide (long-lasting somatostatin ortholog) infusion, and Helicobacter felis infection. GFP(+) cells were also detected in the anterior lobe of the pituitary gland and importantly in the colonic tumor cells induced by administration with azoxymethane and dextran sulfate sodium salt. This transgenic mouse provides a useful tool to study the regulation of mouse gastrin gene in vivo, thus contributing to our understanding of the mechanisms involved in transcriptional control of the gastrin gene.


Gastroenterology | 2013

755 Obesity Accelerates Helicobacter felis-Induced Gastric Carcinogenesis by Enhancing TH17 Responses and Immature Myeloid Cell Trafficking

Russell Ericksen; Shannon Rose; Wataru Shibata; Sureshkumar Muthupalani; James G. Fox; Anthony W. Ferrante; Weiping Han; Timothy C. Wang

Background & Aims: Microbiota has been shown to play a critical role in colorectal cancer (CRC). Most of CRC mucosa-associated Escherichia coli harbor the pks genomic island (pks+ E. coli) responsible for the synthesis of colibactin, a genotoxic compound able to promote colon tumorigenesis in a genetically susceptible mouse model. Our aim was to investigate the mechanisms underlying the tumorigenic activity of E. coli. Methods: Human intestinal epithelial cells HCT116 were infected with pks+ or pksE. coli. Cellular senescence and proliferation were assessed by β-galactosidase activity and MTT assay, respectively. Effect of growth factors secreted from infected cells on the proliferation of uninfected cells was assessed using specific neutralizing antibodies and inhibitors in vitro and in a xenograft mouse model. Human biopsies isolated from CRC tumors colonized with pks+ or pksE. coli were used to validate the experimental data. Results: A transient infection of HCT16 cells with pks+ E. coli increased tumor growth in the xenograft mouse model compared to pksE. coli infection, and the conditioned media derived from pks+ E. coli-infected cells stimulated the growth of uninfected cells. A pretreatment of the conditioned media with an anti-HGF antibody and inhibition of HGF receptor in the xenograft mouse model abolished the pro-proliferative effect of pks+ E. coli, indicating the involvement of the growth factor HGF. HGF was produced by pks+ E. coli-induced senescent cells, which is driven by posttranslational modification of p53 involving small ubiquitin-like modifier (SUMO) peptides. The pks+ E. coli-infected cells displayed a modified pattern of SUMO-conjugated proteins and decreased expression of the SUMO peptidase SENP1, a key regulator of SUMOylation. Over-expression of SENP1 in HCT116 cells abolished the deregulated SUMOylation, cellular senescence, HGF production and the pro-proliferative effect induced by pks+ E. coli infection. Finally, the human CRC tumors colonized by pks+ E. coli expressed a higher level of HGF and decreased SENP1 expression compared to those colonized by pksE. coli, supporting the pathological relevance of the in vitro data. Conclusions: Our study elucidates a new paradigm for the mechanism underlying the carcinogenesis mediated by colibactin-producing bacteria. We propose that those bacteria modulate tumor microenvironment by favoring the emergence of senescent cells, which in turn promote tumor growth by the secretion of growth factors, notably HGF.


Cancer Research | 2012

Abstract 3301: Multiple cytokeratin-19 positive stem cells contribute to the clonal origin of colonic tumors in mice

Samuel Asfaha; Christoph B. Westphalen; Russell Ericksen; Johannes von Burstin; Sarah Stokes; Teresa L. Mastracci; Michael Quante; Wataru Shibata; Anil K. Rustgi; Timothy C. Wang

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Introduction: The clonal origin of tumors is not well understood. Previous studies using chimeric mice have suggested that the origin of tumors in the mammalian small intestine may be polyclonal, with 2 or more progenitor cells contributing to intestinal adenomas. In the intestine, tissue stem cells are believed to be present in small numbers, and either quiescent or actively dividing, as previously shown with Lgr-5+ cells. Cytokeratin 19 (K19) is a potential progenitor marker expressed in the intestinal isthmus. Using the K19 gene promoter to drive Cre recombinase expression, we tracked the lineage of K19+ cells in an inducible fashion. Here, we used K19-CreERT2;ROSA26rLacZ mice to examine whether K19+ cells contribute to colonic tumor formation. Methods: In order to recapitulate the endogenous expression pattern of K19, we used a BAC strategy and generated a transgenic mouse with a Tamoxifen inducible Cre under the control of the K19 promoter (K19-BAC-Cre-ERT2). K19CreERT2 mice were crossed to ROSA26rLacZ (or GFP) reporter mice and treated with tamoxifen (6 mg/ day p.o. for 3 days) to assess K19+ lineage tracing by X-gal or GFP staining. To examine the contribution of the K19+ cells to colonic tumorigenesis, we used the AOM/DSS inflammation-associated model of colonic carcinogenesis. Mice were sacrificed 20 weeks following AOM induction and X-gal staining performed to stain for the K19+ cell lineage. Results: Treatment of control K19CreERT2/Rosa26r mice with tamoxifen resulted in the stochastic labelling of about 20-50% of colonic and intestinal glands, respectively. Consistent with the labeling of stem cells, marked glands stained positive beyond 52 weeks following tamoxifen induction. K19CreERT2/Rosa26r mice treated with AOM or DSS alone following tamoxifen labelling of K19+ glands displayed evidence of K19+ cell expansion and crypt fission, as determined by increased labelling of contiguous X-gal positive glands. K19CreERT2/Rosa26r mice were then treated with a single dose of AOM followed by DSS. When K19+ cells were labelled by tamoxifen administration prior to AOM, we observed that ∼10% tumors were entirely X-gal positive, suggesting derivation from a single K19+ cell. Interestingly, when K19+ cells were labelled by tamoxifen shortly after tumor initiation with AOM, some tumors were derived from both K19 recombined and non-recombined cells in a clonal fashion, suggesting a model whereby a K19+ cell gives rise to multiple K19+ daughter cells that each contribute to the tumor. Conclusions. Using K19CreERT2/Rosa26r mice, we show that K19 marks stem cells within the intestinal isthmus of the small intestine and colon. Using a carcinogen induced model of inflammation-associated carcinogenesis, we demonstrate that K19 expressing cells contribute to tumor formation in a manner consistent with tumor initiation following division of K19+ cells. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3301. doi:1538-7445.AM2012-3301


Cancer Research | 2011

Abstract 5186: Lineage tracing of Cytokeratin 19 labels gastric, intestinal and colonic stem cells

Samuel Asfaha; Russell Ericksen; Johannes von Burstin; Michael Quante; Wataru Shibata; Anil K. Rustgi; Timothy C. Wang

Introduction: Tissue stem cells, characterized by multipotentiality and self renewing ability, are not readily distinguishable from other resident cells. Tissue stem cells are believed to be present in small numbers that may remain quiescent, or divide frequently as previously shown with Lgr-5+ cells. In the gastric antrum, intestine and colon, epithelial cells derived from the stem cells at the crypt base rapidly migrate up towards to the mucosal surface, whereas in the gastric corpus, stem cells located in the isthmus are believed to give rise to epithelial progenitors that undergo bipolar migration and differentiation into pit and oxyntic cell lineages. Cytokeratin 19 (K19) has been shown to be expressed in cells in the gastric isthmus and colonic and intestinal crypts. Therefore, we aimed to use the K19 gene promoter to drive Cre recombinase expression and to track the lineage of K19+ cells in an inducible fashion. Methods: In order to recapitulate the endogenous expression pattern of K19, we used a BAC strategy and generated a transgenic mouse with a Tamoxifen inducible Cre under the control of the K19 promoter (K19-BAC-Cre-ERTM). K19CreERT2 mice were crossed to ROSA26rLacZ (or GFP) reporter mice, treated the mice with tamoxifen (4 mg/ day p.o. for 3 days) and sacrificed at various time points (3 days, 7 days, 26 weeks and 52 weeks) following tamoxifen induction. Results: Treatment of K19CreERT2/Rosa26r mice with tamoxifen resulted in the selective marking of about 30% of gastric and intestinal glands. Following tamoxifen induction, in the corpus a few gastric pit cells near the isthmus were labeled at 3 days, whereas, all of pit cells within the labeled glands stained positively by 7 days. Consistent with the labeling of progenitor or stem cells, all pit cells within marked glands stained positively even up to 52 weeks following tamoxifen induction. In contrast, in the antrum, intestine and colon nearly all epithelial cells were labeled within 3 days following tamoxifen induction. Once again, lineage tracing of all the epithelial cells within marked glands was observed up to 52 weeks following tamoxifen induction, suggesting that K19 likely marks a common progenitor or stem cell in the antrum, intestine and colon. Interestingly, in the small intestine and antrum K19 positive cells do not overlap with Lgr5-GFP+ cells yet clearly demonstrate stem cell properties. Conclusions. Using K19CreERT2/Rosa26r mice, we show that K19 marks pit progenitors in the gastric corpus, as well as stem cells within the antrum, intestine and colon. These findings identify a novel population of stem cells for the lineage tracing of gastric pit cells following gastric injury or cancer, and for tracing epithelial cells in the development of intestinal or colonic cancer. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 5186. doi:10.1158/1538-7445.AM2011-5186


Gastroenterology | 2010

S1695 In Vivo Analysis of Mouse Gastrin Gene Expression by Bac Transgenic EGFP Reporter Mice

Shigeo Takaishi; Wataru Shibata; Hiroyuki Tomita; Guangchun Jin; Xiangdong Yang; Russell Ericksen; Heuijoon Park; Zinaida Dubeikovskaya; Samuel Asfaha; Michael Quante; Kelly S. Betz; Arthur Shulkes; Mildred Yim; Timothy C. Wang

overexpressed Plexin B1 compared to normal pancreas tissues. Endogenous Plexin B1 and Met protein were detected in eight pancreatic cancer cell lines, with a variety of expression levels. Sema 4D stimulation activated Met protein, and significantly induced cell migration. Discussion: Here we show that Sema 4D involves in the regulation of cell motility through its receptor Plexin B1. And the results of immunostaining suggest that Sema 4D could play a role in the interaction between tumor cells and tumor microenvironment in pancreatic cancer tissues. As many attempts are now ongoing to target the tumor stroma, these findings provide a new insight into molecular targeted therapies for pancreatic cancer.


Cancer Research | 2018

Abstract 3477: Metabolic pathway analyses identify proline biosynthesis as a promoter of liver carcinogenesis

Zhaobing Ding; Russell Ericksen; Michael Steckel; Andrea Haegebarth; Sylvia Gruenewald; Weiping Han

Collaboration


Dive into the Russell Ericksen's collaboration.

Top Co-Authors

Avatar

Timothy C. Wang

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Anil K. Rustgi

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James G. Fox

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kelly S. Betz

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge