Network


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

Hotspot


Dive into the research topics where Bobak Parang is active.

Publication


Featured researches published by Bobak Parang.


PLOS ONE | 2013

Dietary selenium deficiency exacerbates DSS-induced epithelial injury and AOM/DSS-induced tumorigenesis.

Caitlyn W. Barrett; Kshipra Singh; Amy K. Motley; Mary K. Lintel; Elena Matafonova; Amber Bradley; Wei Ning; Shenika Poindexter; Bobak Parang; Vishruth K. Reddy; Rupesh Chaturvedi; Barbara Fingleton; Mary Kay Washington; Keith T. Wilson; Sean S. Davies; Kristina E. Hill; Raymond F. Burk; Christopher S. Williams

Selenium (Se) is an essential micronutrient that exerts its functions via selenoproteins. Little is known about the role of Se in inflammatory bowel disease (IBD). Epidemiological studies have inversely correlated nutritional Se status with IBD severity and colon cancer risk. Moreover, molecular studies have revealed that Se deficiency activates WNT signaling, a pathway essential to intestinal stem cell programs and pivotal to injury recovery processes in IBD that is also activated in inflammatory neoplastic transformation. In order to better understand the role of Se in epithelial injury and tumorigenesis resulting from inflammatory stimuli, we examined colonic phenotypes in Se-deficient or -sufficient mice in response to dextran sodium sulfate (DSS)-induced colitis, and azoxymethane (AOM) followed by cyclical administration of DSS, respectively. In response to DSS alone, Se-deficient mice demonstrated increased morbidity, weight loss, stool scores, and colonic injury with a concomitant increase in DNA damage and increases in inflammation-related cytokines. As there was an increase in DNA damage as well as expression of several EGF and TGF-β pathway genes in response to inflammatory injury, we sought to determine if tumorigenesis was altered in the setting of inflammatory carcinogenesis. Se-deficient mice subjected to AOM/DSS treatment to model colitis-associated cancer (CAC) had increased tumor number, though not size, as well as increased incidence of high grade dysplasia. This increase in tumor initiation was likely due to a general increase in colonic DNA damage, as increased 8-OHdG staining was seen in Se-deficient tumors and adjacent, non-tumor mucosa. Taken together, our results indicate that Se deficiency worsens experimental colitis and promotes tumor development and progression in inflammatory carcinogenesis.


Journal of Clinical Investigation | 2015

Selenoprotein P influences colitis-induced tumorigenesis by mediating stemness and oxidative damage

Caitlyn W. Barrett; Vishruth K. Reddy; Sarah P. Short; Amy K. Motley; Mary K. Lintel; Amber Bradley; Tanner J. Freeman; Jefferson Vallance; Wei Ning; Bobak Parang; Shenika Poindexter; Barbara Fingleton; Xi Chen; Mary Kay Washington; Keith T. Wilson; Noah F. Shroyer; Kristina E. Hill; Raymond F. Burk; Christopher S. Williams

Patients with inflammatory bowel disease are at increased risk for colon cancer due to augmented oxidative stress. These patients also have compromised antioxidant defenses as the result of nutritional deficiencies. The micronutrient selenium is essential for selenoprotein production and is transported from the liver to target tissues via selenoprotein P (SEPP1). Target tissues also produce SEPP1, which is thought to possess an endogenous antioxidant function. Here, we have shown that mice with Sepp1 haploinsufficiency or mutations that disrupt either the selenium transport or the enzymatic domain of SEPP1 exhibit increased colitis-associated carcinogenesis as the result of increased genomic instability and promotion of a protumorigenic microenvironment. Reduced SEPP1 function markedly increased M2-polarized macrophages, indicating a role for SEPP1 in macrophage polarization and immune function. Furthermore, compared with partial loss, complete loss of SEPP1 substantially reduced tumor burden, in part due to increased apoptosis. Using intestinal organoid cultures, we found that, compared with those from WT animals, Sepp1-null cultures display increased stem cell characteristics that are coupled with increased ROS production, DNA damage, proliferation, decreased cell survival, and modulation of WNT signaling in response to H2O2-mediated oxidative stress. Together, these data demonstrate that SEPP1 influences inflammatory tumorigenesis by affecting genomic stability, the inflammatory microenvironment, and epithelial stem cell functions.


Gut | 2017

BVES regulates c-Myc stability via PP2A and suppresses colitis-induced tumourigenesis

Bobak Parang; Andrew M. Kaz; Caitlyn W. Barrett; Sarah P. Short; Wei Ning; Cody Keating; Mukul K. Mittal; Rishi D. Naik; Mary Kay Washington; Frank Revetta; J. Joshua Smith; Xi Chen; Keith T. Wilson; Thomas Brand; David M. Bader; William P. Tansey; Ru Chen; Teresa A. Brentnall; William M. Grady; Christopher S. Williams

Objective Blood vessel epicardial substance (BVES) is a tight junction-associated protein that regulates epithelial-mesenchymal states and is underexpressed in epithelial malignancy. However, the functional impact of BVES loss on tumourigenesis is unknown. Here we define the in vivo role of BVES in colitis-associated cancer (CAC), its cellular function and its relevance to patients with IBD. Design We determined BVES promoter methylation status using an Infinium HumanMethylation450 array screen of patients with UC with and without CAC. We also measured BVES mRNA levels in a tissue microarray consisting of normal colons and CAC samples. Bves−/− and wild-type mice (controls) were administered azoxymethane (AOM) and dextran sodium sulfate (DSS) to induce tumour formation. Last, we used a yeast two-hybrid screen to identify BVES interactors and performed mechanistic studies in multiple cell lines to define how BVES reduces c-Myc levels. Results BVES mRNA was reduced in tumours from patients with CAC via promoter hypermethylation. Importantly, BVES promoter hypermethylation was concurrently present in distant non-malignant-appearing mucosa. As seen in human patients, Bves was underexpressed in experimental inflammatory carcinogenesis, and Bves−/− mice had increased tumour multiplicity and degree of dysplasia after AOM/DSS administration. Molecular analysis of Bves−/− tumours revealed Wnt activation and increased c-Myc levels. Mechanistically, we identified a new signalling pathway whereby BVES interacts with PR61α, a protein phosphatase 2A regulatory subunit, to mediate c-Myc destruction. Conclusion Loss of BVES promotes inflammatory tumourigenesis through dysregulation of Wnt signalling and the oncogene c-Myc. BVES promoter methylation status may serve as a CAC biomarker.


Methods of Molecular Biology | 2016

AOM/DSS Model of Colitis-Associated Cancer

Bobak Parang; Caitlyn W. Barrett; Christopher S. Williams

Our understanding of colitis-associated carcinoma (CAC) has benefited substantially from mouse models that faithfully recapitulate human CAC. Chemical models, in particular, have enabled fast and efficient analysis of genetic and environmental modulators of CAC without the added requirement of time-intensive genetic crossings. Here we describe the Azoxymethane (AOM)/Dextran Sodium Sulfate (DSS) mouse model of inflammatory colorectal cancer.


The FASEB Journal | 2015

The transcriptional corepressor MTGR1 regulates intestinal secretory lineage allocation

Bobak Parang; Daniel O. Rosenblatt; Amanda D. Williams; Mary Kay Washington; Frank Revetta; Sarah P. Short; Vishruth K. Reddy; Aubrey Hunt; Noah F. Shroyer; Michael E. Engel; Scott W. Hiebert; Christopher S. Williams

Notch signaling largely determines intestinal epithelial cell fate. High Notch activity drives progenitors toward absorptive enterocytes by repressing secretory differentiation programs, whereas low Notch permits secretory cell assignment. Myeloid translocation gene‐related 1 (MTGR1) is a transcriptional corepressor in the myeloid translocation gene/Eight‐Twenty‐One family. Given that Mtgr1–/– mice have a dramatic reduction of intestinal epithelial secretory cells, we hypothesized that MTGR1 is a key repressor of Notch signaling. In support of this, transcriptome analysis of laser capture microdissected Mtgr1–/– intestinal crypts revealed Notch activation, and secretory markers Mucin2, Chromogranin A, and Growth factor‐independent 1 (Gfi1) were down‐regulated in Mtgr1–/– whole intestines and Mtgr1–/– enteroids. We demonstrate that MTGR1 is in a complex with Suppressor of Hairless Homolog, a key Notch effector, and represses Notch‐induced Hairy/Enhancer of Split 1 activity. Moreover, pharmacologic Notch inhibition using a γ‐secretase inhibitor (GSI) rescued the hyperproliferative baseline phenotype in the Mtgr1–/– intestine and increased production of goblet and enteroendocrine lineages in Mtgr1–/– mice. GSI increased Paneth cell production in wild‐type mice but failed to do so in Mtgr1–/– mice. We determined that MTGR1 can interact with GFI1, a transcriptional corepressor required for Paneth cell differentiation, and repress GFI1 targets. Overall, the data suggest that MTGR1, a transcriptional corepressor well characterized in hematopoiesis, plays a critical role in intestinal lineage allocation.—Parang, B., Rosenblatt, D., Williams, A. D., Washington, M. K., Revetta, F., Short, S. P., Reddy, V. K., Hunt, A., Shroyer, N. F., Engel, M. E., Hiebert, S. W., Williams, C. S., The transcriptional corepressors MTGR1 regulates intestinal secretory lineage allocation. FASEB J. 29, 786–795 (2015). www.fasebj.org


Oncogene | 2016

Myeloid translocation genes differentially regulate colorectal cancer programs

Bobak Parang; Amber Bradley; Mukul K. Mittal; Sarah P. Short; Joshua J. Thompson; Caitlyn W. Barrett; Rishi D. Naik; A J Bilotta; Mary Kay Washington; Frank Revetta; Jesse J. Smith; Xi Chen; Keith T. Wilson; Scott W. Hiebert; Christopher S. Williams

Myeloid translocation genes (MTGs), originally identified as chromosomal translocations in acute myelogenous leukemia, are transcriptional corepressors that regulate hematopoietic stem cell programs. Analysis of The Cancer Genome Atlas (TCGA) database revealed that MTGs were mutated in epithelial malignancy and suggested that loss of function might promote tumorigenesis. Genetic deletion of MTGR1 and MTG16 in the mouse has revealed unexpected and unique roles within the intestinal epithelium. Mtgr1−/− mice have progressive depletion of all intestinal secretory cells, and Mtg16−/− mice have a decrease in goblet cells. Furthermore, both Mtgr1−/− and Mtg16−/− mice have increased intestinal epithelial cell proliferation. We thus hypothesized that loss of MTGR1 or MTG16 would modify Apc1638/+-dependent intestinal tumorigenesis. Mtgr1−/− mice, but not Mtg16−/− mice, had a 10-fold increase in tumor multiplicity. This was associated with more advanced dysplasia, including progression to invasive adenocarcinoma, and augmented intratumoral proliferation. Analysis of chromatin immunoprecipitation sequencing data sets for MTGR1 and MTG16 targets indicated that MTGR1 can regulate Wnt and Notch signaling. In support of this, immunohistochemistry and gene expression analysis revealed that both Wnt and Notch signaling pathways were hyperactive in Mtgr1−/− tumors. Furthermore, in human colorectal cancer (CRC) samples MTGR1 was downregulated at both the transcript and protein level. Overall our data indicates that MTGR1 has a context-dependent effect on intestinal tumorigenesis.


Mucosal Immunology | 2018

BVES is required for maintenance of colonic epithelial integrity in experimental colitis by modifying intestinal permeability

Yash A. Choksi; Vishruth K. Reddy; Kshipra Singh; Caitlyn W. Barrett; Sarah P. Short; Bobak Parang; Cody Keating; Joshua J. Thompson; Thomas Verriere; Rachel E. Brown; M. Blanca Piazuelo; David M. Bader; M. Kay Washington; Mukul K. Mittal; Thomas Brand; Alain P. Gobert; Lori A. Coburn; Keith T. Wilson; Christopher S. Williams

Blood vessel epicardial substance (BVES), or POPDC1, is a tight junction-associated transmembrane protein that modulates epithelial-to-mesenchymal transition (EMT) via junctional signaling pathways. There have been no in vivo studies investigating the role of BVES in colitis. We hypothesized that BVES is critical for maintaining colonic epithelial integrity. At baseline, Bves−/− mouse colons demonstrate increased crypt height, elevated proliferation, decreased apoptosis, altered intestinal lineage allocation, and dysregulation of tight junctions with functional deficits in permeability and altered intestinal immunity. Bves−/− mice inoculated with Citrobacter rodentium had greater colonic injury, increased colonic and mesenteric lymph node bacterial colonization, and altered immune responses after infection. We propose that increased bacterial colonization and translocation result in amplified immune responses and worsened injury. Similarly, dextran sodium sulfate (DSS) treatment resulted in greater histologic injury in Bves−/− mice. Two different human cell lines (Caco2 and HEK293Ts) co-cultured with enteropathogenic E. coli showed increased attaching/effacing lesions in the absence of BVES. Finally, BVES mRNA levels were reduced in human ulcerative colitis (UC) biopsy specimens. Collectively, these studies suggest that BVES plays a protective role both in ulcerative and infectious colitis and identify BVES as a critical protector of colonic mucosal integrity.


JCI insight | 2017

MTG16 is a tumor suppressor in colitis-associated carcinoma

Elizabeth McDonough; Caitlyn W. Barrett; Bobak Parang; Mukul K. Mittal; Jesse J. Smith; Amber Bradley; Yash A. Choksi; Lori A. Coburn; Sarah P. Short; Joshua J. Thompson; Baolin Zhang; Shenika Poindexter; Melissa A. Fischer; Xi Chen; Li J; Frank Revetta; Rishi D. Naik; Mary Kay Washington; Michael J. Rosen; Scott W. Hiebert; Keith T. Wilson; Christopher S. Williams

MTG16 is a member of the myeloid translocation gene (MTG) family of transcriptional corepressors. While MTGs were originally identified in chromosomal translocations in acute myeloid leukemia, recent studies have uncovered a role in intestinal biology. For example, Mtg16-/- mice have increased intestinal proliferation and are more sensitive to intestinal injury in colitis models. MTG16 is also underexpressed in patients with moderate/severe ulcerative colitis. Based on these findings, we postulated that MTG16 might protect against colitis-associated carcinogenesis. MTG16 was downregulated at the protein and RNA levels in patients with inflammatory bowel disease and in those with colitis-associated carcinoma. Mtg16-/- mice subjected to inflammatory carcinogenesis modeling exhibited worse colitis and increased tumor multiplicity and size. Loss of MTG16 also increased severity of dysplasia, apoptosis, proliferation, DNA damage, and WNT signaling. Moreover, transplantation of WT marrow into Mtg16-/- mice failed to rescue the Mtg16-/- protumorigenic phenotypes, indicating an epithelium-specific role for MTG16. While MTG dysfunction is widely appreciated in hematopoietic malignancies, the role of this gene family in epithelial homeostasis, and in colon cancer, was unrealized. This report identifies MTG16 as an important modulator of colitis and tumor development in inflammatory carcinogenesis.


Medical Education Online | 2016

A workshop on leadership for senior MD-PhD students.

Catherine B. Meador; Bobak Parang; Melissa A. Musser; Rachana Haliyur; David A. Owens; Terence S. Dermody

Leadership skills are essential for a successful career as a physician-scientist, yet many MD–PhD training programs do not offer formal training in leadership. The Vanderbilt Medical Scientist Training Program (MSTP) previously established a 2-day leadership workshop that has been held biennially since 2006 for students in the first and second years of the graduate school portion of combined MD and PhD training (G1/G2 students). Workshop attendees have consistently rated this workshop as a highly effective experience. However, opportunities for structured training in leadership competencies during the subsequent 3–5 years of MD–PhD training are limited. Given the success of the G1/G2 leadership workshop and the need for continuity in this model of leadership training, we developed a half-day workshop for MSTP students in the clinical years of medical school (M3/M4 students) to foster continued training in leadership. Our workshop curriculum, based in part on original cases drafted by Vanderbilt MSTP students, provides concrete strategies to manage conflict and navigate leadership transitions in the physician-scientist career path. The curriculum emphasizes both short-term competencies, such as effective participation as a member of a clinical team, and long-term competencies, such as leadership of a research team, division, or department. Our inaugural senior leadership workshop, held in August, 2015, was judged by student participants to be well organized and highly relevant to leadership concepts and skills. It will be offered biennially in our training curriculum for M3 and M4 MSTP students.Leadership skills are essential for a successful career as a physician-scientist, yet many MD-PhD training programs do not offer formal training in leadership. The Vanderbilt Medical Scientist Training Program (MSTP) previously established a 2-day leadership workshop that has been held biennially since 2006 for students in the first and second years of the graduate school portion of combined MD and PhD training (G1/G2 students). Workshop attendees have consistently rated this workshop as a highly effective experience. However, opportunities for structured training in leadership competencies during the subsequent 3-5 years of MD-PhD training are limited. Given the success of the G1/G2 leadership workshop and the need for continuity in this model of leadership training, we developed a half-day workshop for MSTP students in the clinical years of medical school (M3/M4 students) to foster continued training in leadership. Our workshop curriculum, based in part on original cases drafted by Vanderbilt MSTP students, provides concrete strategies to manage conflict and navigate leadership transitions in the physician-scientist career path. The curriculum emphasizes both short-term competencies, such as effective participation as a member of a clinical team, and long-term competencies, such as leadership of a research team, division, or department. Our inaugural senior leadership workshop, held in August, 2015, was judged by student participants to be well organized and highly relevant to leadership concepts and skills. It will be offered biennially in our training curriculum for M3 and M4 MSTP students.


Gastroenterology | 2014

717 Loss of MTGR1 Accelerates Intestinal Tumor Burden

Bobak Parang; Amber Bradley; Mukul K. Mittal; Kay Washington; Frank Revetta; Anthony Bilotta; J. Joshua Smith; Xi Chen; Noah F. Shroyer; Christopher S. Williams

Recent genome-wide association studies have uncovered genetic links for a number of complex diseases, but these powerful tools have not yet been widely applied to the clinical management of disease. Here we describe our experience using a genetic approach to uncover newmolecular insights in a young patient presenting with unexplained infections and chronic intestinal inflammation whomwe eventually diagnosed with anti-goblet cell antibody positive autoimmune enterocolopathy. Using a single-nucleotide polymorphism (SNP) array, we identified a deletion of the gene Nuclear Factor of Activated T cells-5 (NFAT5) in one chromosome in the patient and verified this deficiency at the mRNA and protein levels. In mice, NFAT5 has previously been shown to regulate cellular responses to osmotic stress and to play a critical role in influencing differentiation of pro-inflammatory T helper 17 (Th17) lymphocytes. We demonstrate for the first time that human NFAT5 deficiency appears to result in functional defects in both innate and adaptive immunity, with a markedly reduced number of natural killer (NK) cells, and impaired proliferation and cytokine production by T lymphocytes. We confirm the molecular link between immune cell function and NFAT5 deficiency using human cells as well as cells from mice deficient in NFAT5. Together our approach and observations demonstrate the potential power of genetic analyses to facilitate clinical diagnosis and to provide novel mechanistic insights into disease.

Collaboration


Dive into the Bobak Parang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wei Ning

Vanderbilt University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Keith T. Wilson

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kay Washington

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge