Network


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

Hotspot


Dive into the research topics where Bogdan C. Paun is active.

Publication


Featured researches published by Bogdan C. Paun.


Hepatology | 2009

MicroRNA-21 is overexpressed in human cholangiocarcinoma and regulates programmed cell death 4 and tissue inhibitor of metalloproteinase 3†

Florin M. Selaru; Alexandru Olaru; Takatsugu Kan; Stefan David; Yulan Cheng; Yuriko Mori; Jian Yang; Bogdan C. Paun; Zhe Jin; Rachana Agarwal; James P. Hamilton; John A. Abraham; Christos S. Georgiades; Hector Alvarez; Perumal Vivekanandan; Wayne Yu; Anirban Maitra; Michael Torbenson; Paul J. Thuluvath; Gregory J. Gores; Nicholas F. LaRusso; Ralph H. Hruban; Stephen J. Meltzer

Cholangiocarcinomas (CCAs) are aggressive cancers, with high mortality and poor survival rates. Only radical surgery offers patients some hope of cure; however, most patients are not surgical candidates because of late diagnosis secondary to relatively poor accuracy of diagnostic means. MicroRNAs (miRs) are involved in every cancer examined, but they have not been evaluated in primary CCA. In this study, miR arrays were performed on five primary CCAs and five normal bile duct specimens (NBDs). Several miRs were dysregulated and miR‐21 was overexpressed in CCAs. miR‐21 differential expression in these 10 specimens was verified by quantitative reverse transcriptase polymerase chain reaction (qRT‐PCR). To validate these findings, qRT‐PCR for miR‐21 was then performed on 18 additional primary CCAs and 12 normal liver specimens. MiR‐21 was 95% sensitive and 100% specific in distinguishing between CCA and normal tissues, with an area under the receiver operating characteristic curve of 0.995. Inhibitors of miR‐21 increased protein levels of programmed cell death 4 (PDCD4) and tissue inhibitor of metalloproteinases 3 (TIMP3). Notably, messenger RNA levels of TIMP3 were significantly lower in CCAs than in normals. Conclusions: MiR‐21 is overexpressed in human CCAs. Furthermore, miR‐21 may be oncogenic, at least in part, by inhibiting PDCD4 and TIMP3. Finally, these data suggest that TIMP3 is a candidate tumor suppressor gene in the biliary tree. (HEPATOLOGY 2009.)


Cancer Research | 2009

A multicenter, double-blinded validation study of methylation biomarkers for progression prediction in Barrett’s esophagus

Zhe Jin; Yulan Cheng; Wen Gu; Yingye Zheng; Fumiaki Sato; Yuriko Mori; Alexandru Olaru; Bogdan C. Paun; Jian Yang; Takatsugu Kan; Tetsuo Ito; James P. Hamilton; Florin M. Selaru; Rachana Agarwal; Stefan David; John M. Abraham; Herbert C. Wolfsen; Michael B. Wallace; Nicholas J. Shaheen; Kay Washington; Jean Q Wang; Marcia I. Canto; Achyut K. Bhattacharyya; Mark A. Nelson; Paul A. Wagner; Yvonne Romero; Kenneth K. Wang; Ziding Feng; Richard E. Sampliner; Stephen J. Meltzer

Esophageal adenocarcinoma risk in Barretts esophagus (BE) is increased 30- to 125-fold versus the general population. Among all BE patients, however, neoplastic progression occurs only once per 200 patient-years. Molecular biomarkers are therefore needed to risk-stratify patients for more efficient surveillance endoscopy and to improve the early detection of progression. We therefore performed a retrospective, multicenter, double-blinded validation study of eight BE progression prediction methylation biomarkers. Progression or nonprogression were determined at 2 years (tier 1) and 4 years (tier 2). Methylation was assayed in 145 nonprogressors and 50 progressors using real-time quantitative methylation-specific PCR. Progressors were significantly older than nonprogressors (70.6 versus 62.5 years; P < 0.001). We evaluated a linear combination of the eight markers, using coefficients from a multivariate logistic regression analysis. Areas under the ROC curve (AUC) were high in the 2-year, 4-year, and combined data models (0.843, 0.829, and 0.840; P < 0.001, <0.001, and <0.001, respectively). In addition, even after rigorous overfitting correction, the incremental AUCs contributed by panels based on the 8 markers plus age versus age alone were substantial (Delta-AUC = 0.152, 0.114, and 0.118, respectively) in all 3 models. A methylation biomarker-based panel to predict neoplastic progression in BE has potential clinical value in improving both the efficiency of surveillance endoscopy and the early detection of neoplasia.


Oncogene | 2006

Transcriptional profiling suggests that Barrett's metaplasia is an early intermediate stage in esophageal adenocarcinogenesis

Suna Wang; M Zhan; Jing Yin; John M. Abraham; Yuriko Mori; Fumiako Sato; Yan Xu; Andreea Olaru; A T Berki; H Li; Karsten Schulmann; Takatsugu Kan; James P. Hamilton; Bogdan C. Paun; M M Yu; Zhe Jin; Yulan Cheng; Tetsuo Ito; C Mantzur; Bruce D. Greenwald; Stephen J. Meltzer

To investigate the relationship between Barretts esophagus (BE) and esophageal adenocarcinoma (EAC), we determined gene expression profiles of discrete pathological stages of esophageal neoplasia using a sequence-verified human cDNA microarray. Fifty one RNAs, comprising 24 normal esophagi (NE), 18 BEs, and nine EACs were hybridized to cDNA microarrays. Five statistical analyses were used for the data analysis. Genes showing significantly different expression levels among the three sample groups were identified. Genes were grouped into functional categories based on the Gene Ontology Consortium. Surprisingly, the expression pattern of BE was significantly more similar to EAC than to NE, notwithstanding the known histopathologic differences between BE and EAC. The pattern of NE was clearly distinct from that of EAC. Thirty-six genes were the most differentially modulated, according to these microarray data, in BE-associated neoplastic progression. Twelve genes were significantly differentially expressed in cancer-associated BEs plus EAC (as a single combined tissue group) vs noncancer-associated BEs. These genes represent potential biomarkers to diagnose EAC at its early stages. Our results demonstrate that molecular events at the transcriptional level in BE are remarkably similar to BEs-associated adenocarcinoma of the esophagus. This finding alarmingly implies that BE is biologically closer to cancer than to normal esophagus, and that the cancer risk of BE is perhaps higher than we had imagined. These findings suggest that changes modulated at the molecular biologic level supervene earlier than histologic changes, and that BE is an early intermediate stage in the process of EAC.


Inflammatory Bowel Diseases | 2011

Dynamic changes in the expression of microRNA-31 during inflammatory bowel disease-associated neoplastic transformation

Alexandru Olaru; Florin M. Selaru; Yuriko Mori; Christine Vazquez; Stefan David; Bogdan C. Paun; Yulan Cheng; Zhe Jin; Jian Yang; Rachana Agarwal; John M. Abraham; Themistocles Dassopoulos; Mary L. Harris; Theodore M. Bayless; John H. Kwon; Noam Harpaz; Ferenc Livak; Stephen J. Meltzer

Background: Patients with inflammatory bowel disease (IBD) are at increased risk of developing colorectal cancer. Aberrant microRNA (miR) expression has been linked to carcinogenesis; however, no reports document a relationship between IBD‐related neoplasia (IBDN) and altered miR expression. In the current study we sought to identify specific miR dysregulation along the normal–inflammation–cancer axis. Methods: miR microarrays and quantitative reverse‐transcriptase polymerase chain reaction (RT‐PCR) were used to detect dysregulated miRs. Receiver operating characteristic curve analysis was employed to test for potential usefulness of miR‐31 as a disease marker of IBDNs. In silico prediction analysis, Western blot, and luciferase activity measurement were employed for target identification. Results: Several dysregulated miRs were identified between chronically inflamed mucosae and dysplasia arising in IBD. MiR‐31 expression increases in a stepwise fashion during progression from normal to IBD to IBDN and accurately discriminated IBDNs from normal or chronically inflamed tissues in IBD patients. Finally, we identified factor inhibiting hypoxia inducible factor 1 as a direct target of miR‐31. Conclusions: Our study reveals specific miR dysregulation as chronic inflammation progresses to dysplasia. MiR‐31 expression levels increase with disease progression and accurately discriminates between distinct pathological entities that coexist in IBD patients. The novel effect of miR‐31 on regulating factor inhibiting hypoxia inducible factor 1 expression provides a new insight on the pathogenesis of IBDN. (Inflamm Bowel Dis 2011;)


Clinical Cancer Research | 2007

Hypermethylation of tachykinin-1 is a potential biomarker in human esophageal cancer.

Zhe Jin; Alexandru Olaru; Jian Yang; Fumiaki Sato; Yulan Cheng; Takatsugu Kan; Yuriko Mori; Carmit Mantzur; Bogdan C. Paun; James P. Hamilton; Tetsuo Ito; Suna Wang; Stefan David; Rachana Agarwal; David G. Beer; John M. Abraham; Stephen J. Meltzer

Purpose: Our aim was to investigate whether and at what stage hypermethylation of the tachykinin-1 (TAC1) gene is associated with human esophageal neoplastic transformation. Experimental Design:TAC1 promoter hypermethylation was examined by real-time methylation-specific PCR in 258 human esophageal specimens and 126 plasma samples from patients or tissues at various stages of neoplastic evolution. Results:TAC1 hypermethylation in tissue samples showed highly discriminative receiver-operator characteristic curve profiles, clearly distinguishing esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC) from normal esophagus (P < 0.0001). Both frequencies and normalized methylation values of TAC1 tissue methylation were significantly higher in Barretts metaplasia (BE), dysplastic Barretts esophagus, EAC, and ESCC than in normal esophagus (P < 0.01). The frequency of TAC1 hypermethylation increased dramatically and early during neoplastic progression, from 7.5% in normal esophagus to 55.6% in BE from patients with Barretts metaplasia alone, 57.5% in dysplastic Barretts esophagus, and 61.2% in EAC. There was a significant relationship between TAC1 hypermethylation and BE segment length, a known clinical risk factor for neoplastic progression. Twelve (50%) of 24 ESCC exhibited TAC1 hypermethylation. Overall patient survival correlated significantly with TAC1 methylation status in ESCC patients (mean survival, 22 versus 110 months; P = 0.0102, log-rank test), but not in EAC patients. Both mean normalized methylation values and frequency of TAC1 hypermethylation in plasma samples were significantly higher in EAC patients than in control subjects. Treatment of KYSE220 ESCC and BIC EAC cells with 5-aza-2′-deoxycytidine reduced TAC1 methylation and increased TAC1 mRNA expression. Conclusions:TAC1 promoter hypermethylation is a common event in both major histologic types of human esophageal carcinoma, occurs early, correlates with other progression risk factors in esophageal adenocarcinogenesis, and is a tissue biomarker of a poor prognosis in ESCC. Circulating methylated TAC1 promoter DNA also offers potential as a biomarker for the diagnosis of EAC.


PLOS ONE | 2008

Three-tiered risk stratification model to predict progression in Barrett's esophagus using epigenetic and clinical features

Fumiaki Sato; Zhe Jin; Karsten Schulmann; Jean Wang; Bruce D. Greenwald; Tetsuo Ito; Takatsugu Kan; James P. Hamilton; Jian Yang; Bogdan C. Paun; Stefan David; Alexandru Olaru; Yulan Cheng; Yuriko Mori; John M. Abraham; Harris G. Yfantis; Tsung Teh Wu; Mary Fredericksen; Kenneth K. Wang; Marcia I. Canto; Yvonne Romero; Ziding Feng; Stephen J. Meltzer

Background Barretts esophagus predisposes to esophageal adenocarcinoma. However, the value of endoscopic surveillance in Barretts esophagus has been debated because of the low incidence of esophageal adenocarcinoma in Barretts esophagus. Moreover, high inter-observer and sampling-dependent variation in the histologic staging of dysplasia make clinical risk assessment problematic. In this study, we developed a 3-tiered risk stratification strategy, based on systematically selected epigenetic and clinical parameters, to improve Barretts esophagus surveillance efficiency. Methods and Findings We defined high-grade dysplasia as endpoint of progression, and Barretts esophagus progressor patients as Barretts esophagus patients with either no dysplasia or low-grade dysplasia who later developed high-grade dysplasia or esophageal adenocarcinoma. We analyzed 4 epigenetic and 3 clinical parameters in 118 Barretts esophagus tissues obtained from 35 progressor and 27 non-progressor Barretts esophagus patients from Baltimore Veterans Affairs Maryland Health Care Systems and Mayo Clinic. Based on 2-year and 4-year prediction models using linear discriminant analysis (area under the receiver-operator characteristic (ROC) curve: 0.8386 and 0.7910, respectively), Barretts esophagus specimens were stratified into high-risk (HR), intermediate-risk (IR), or low-risk (LR) groups. This 3-tiered stratification method retained both the high specificity of the 2-year model and the high sensitivity of the 4-year model. Progression-free survivals differed significantly among the 3 risk groups, with p = 0.0022 (HR vs. IR) and p<0.0001 (HR or IR vs. LR). Incremental value analyses demonstrated that the number of methylated genes contributed most influentially to prediction accuracy. Conclusions This 3-tiered risk stratification strategy has the potential to exert a profound impact on Barretts esophagus surveillance accuracy and efficiency.


Oncogene | 2007

Hypermethylation of the nel-like 1 gene is a common and early event and is associated with poor prognosis in early-stage esophageal adenocarcinoma

Zhe Jin; Yuriko Mori; Jr Ming Yang; Fumiaki Sato; Tetsuo Ito; Yulan Cheng; Bogdan C. Paun; James P. Hamilton; Takatsugu Kan; Alexandru Olaru; Stefan David; Rachana Agarwal; John M. Abraham; David G. Beer; Elizabeth A. Montgomery; Stephen J. Meltzer

The nel-like1 (NELL1) gene maps to chromosome 11p15, which frequently undergoes loss of heterozygosity in esophageal adenocarcinoma (EAC). NELL1 promoter hypermethylation was examined by real-time methylation-specific polymerase chain reaction in 259 human esophageal tissues. Hypermethylation of this promoter showed highly discriminative receiver–operator characteristic curve profiles, clearly distinguishing esophageal squamous cell carcinoma (ESCC) and EAC from normal esophagus (NE) (P<0.001). NELL1 normalized methylation values were significantly higher in Barretts metaplasia (BE), dysplastic Barretts (D) and EAC than in NE (P<0.0000001). NELL1 hypermethylation frequency was zero in NE but increased early during neoplastic progression, to 41.7% in BE from patients with Barretts alone, 52.5% in D and 47.8% in EAC. There was a significant correlation between NELL1 hypermethylation and BE segment length. Three (11.5%) of 26 ESCCs exhibited NELL1 hypermethylation. Survival correlated inversely with NELL1 hypermethylation in patients with stages I–II (P=0.0264) but not in stages III–IV (P=0.68) EAC. Treatment of KYSE220 ESCC and BIC EAC cells with 5-aza-2′-deoxycytidine reduced NELL1 methylation and increased NELL1 mRNA expression. NELL1 mRNA levels in EACs with an unmethylated NELL1 promoter were significantly higher than those in EACs with a methylated promoter (P=0.02). Promoter hypermethylation of NELL1 is a common, tissue-specific event in human EAC, occurs early during Barretts-associated esophageal neoplastic progression, and is a potential biomarker of poor prognosis in early-stage EAC.


Clinical Cancer Research | 2006

Reprimo Methylation Is a Potential Biomarker of Barrett's-Associated Esophageal Neoplastic Progression

James P. Hamilton; Fumiaki Sato; Zhe Jin; Bruce D. Greenwald; Tetsuo Ito; Yuriko Mori; Bogdan C. Paun; Takatsugu Kan; Yulan Cheng; Suna Wang; Jian Yang; John M. Abraham; Stephen J. Meltzer

Purpose:Reprimo, a candidate tumor-suppressor gene, regulates p53-mediated cell cycle arrest at G2 phase, and tumor-suppressor gene methylation is involved in the pathogenesis and progression of esophageal cancer. Our aim was to determine whether and at what phase of neoplastic progression Reprimo methylation occurs in Barretts adenocarcinogenesis, as well as its columnar or squamous cell-type specificity. We also sought to determine whether Reprimo expression could be restored in vitro by the demethylating agent 5-aza-deoxycytidine (5AzaC). Experimental Design: Quantitative methylation-specific PCR for Reprimo was done using an ABI7700 (Taqman) apparatus on 175 endoscopic biopsy specimens. In addition, reverse transcription-PCR and quantitative methylation-specific PCR were done on esophageal carcinoma cells before and after treatment with 5AzaC. Results: In Barretts esophagus (BE; P = 0.001), high-grade dysplasia (HGD; P = 0.001), and esophageal adenocarcinoma (EAC; P = 0.00003), the level and frequency of Reprimo methylation were significantly higher than in normal esophagus (NE). There was no statistically significant difference between BE and EAC, HGD and EAC, or NE and esophageal squamous cell carcinoma (ESCC). Reprimo methylation occurred in 0 of 19 NE samples, 6 (13%) of 45 ESCC, 9 (36%) of 25 BE, 7 (64%) of 11 HGD, and 47 (63%) of 75 EAC. Analysis of Reprimo methylation in EAC versus NE revealed an area under the receiver-operator characteristic curve of 0.812 (P < 0.00001; 95% confidence interval, 0.73-0.90). In vitro 5AzaC treatment of OE33 EAC cells reduced Reprimo methylation and increased Reprimo expression. Conclusions:Reprimo methylation occurs significantly more frequently in BE, HGD, and EAC than in NE or ESCC, suggesting that this epigenetic alteration is a specialized columnar, cell-specific early event with potential as a biomarker for the early detection of esophageal neoplasia.


International Journal of Cancer | 2008

Promoter hypermethylation of CDH13 is a common, early event in human esophageal adenocarcinogenesis and correlates with clinical risk factors

Zhe Jin; Yulan Cheng; Alexandru Olaru; Takatsugu Kan; Jian Yang; Bogdan C. Paun; Tetsuo Ito; James P. Hamilton; Stefan David; Rachana Agarwal; Florin M. Selaru; Fumiaki Sato; John M. Abraham; David G. Beer; Yuriko Mori; Yutaka Shimada; Stephen J. Meltzer

Although the CDH13 gene has been shown to undergo epigenetic silencing by promoter methylation in many types of tumors, hypermethylation of this gene in Barretts‐associated esophageal adenocarcinogenesis has not been studied. Two hundred fifty‐nine human esophageal tissues were therefore examined for CDH13 promoter hypermethylation by real‐time methylation‐specific PCR. CDH13 hypermethylation showed discriminative receiver‐operator characteristic curve profiles, sharply demarcating esophageal adenocarcinoma (EAC) from esophageal squamous cell carcinoma (ESCC) and normal esophagus (NE) (p < 0.0001). CDH13 normalized methylation values (NMV) were significantly higher in Barretts esophagus (BE), dysplastic BE (D) and EAC than in NE (p < 0.0000001). CDH13 hypermethylation frequency was 0% in NE but increased early during neoplastic progression, rising to 70% in BE, 77.5% in D and 76.1% in EAC. Both CDH13 hypermethylation frequency and its mean NMV were significantly higher in BE with than without accompanying EAC. In contrast, only 5 (19.2%) of 26 ESCCs exhibited CDH13 hypermethylation. Furthermore, both CDH13 hypermethylation frequency and its mean NMV were significantly higher in EAC than in ESCC, as well as in BE or D vs. ESCC. Interestingly, mean CDH13 NMV was significantly lower in short‐segment than in long‐segment BE, a known clinical risk factor for neoplastic progression. Similarly, BE segment length was significantly lower in specimens with unmethylated than with methylated CDH13 promoters. 5‐aza‐2′‐deoxycytidine treatment of OE33 EAC and KYSE220 ESCC cells reduced CDH13 methylation and increased CDH13 mRNA expression. These findings suggest that hypermethylation of CDH13 is a common, tissue‐specific event in human EAC, occurs early during BE‐associated neoplastic progression, and correlates with known clinical neoplastic progression risk factors.


Cancer | 2010

Relation Between Normal Rectal Methylation, Smoking Status, and the Presence or Absence of Colorectal Adenomas

Bogdan C. Paun; Debra Kukuruga; Zhe Jin; Yuriko Mori; Yulan Cheng; Mark D. Duncan; Sanford A. Stass; Elizabeth A. Montgomery; David F. Hutcheon; Stephen J. Meltzer

Colorectal cancer (CRC) is 1 of the leading causes of death in the Western world. CRC develops from premalignant lesions, chiefly colorectal adenomas. Currently, the most accurate and recommended screening method for finding colorectal adenomas is colonoscopy performed on all individuals aged >50 years. However, the costs and risks associated with this procedure are relatively high. The objectives of the current study were to correlate epigenetic alterations that occur in normal rectal mucosa, smoking status, and age with the presence or absence of concomitant colorectal adenomas and to assess the potential clinical value of methylation in normal rectal biopsies as a screening assay for the presence of polyps and, hence, the need for a full colonoscopy.

Collaboration


Dive into the Bogdan C. Paun's collaboration.

Top Co-Authors

Avatar

Stephen J. Meltzer

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yuriko Mori

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Yulan Cheng

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Zhe Jin

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stefan David

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

James P. Hamilton

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

John M. Abraham

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jian Yang

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge