Network


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

Hotspot


Dive into the research topics where Denada Dibra is active.

Publication


Featured researches published by Denada Dibra.


Cancer Research | 2014

Universal Marker and Detection Tool for Human Sarcoma Circulating Tumor Cells

Arun Satelli; Abhisek Mitra; Jeffry Cutrera; Marcos Devarie; Xueqing Xia; Davis R. Ingram; Denada Dibra; Neeta Somaiah; Keila E. Torres; Vinod Ravi; Joseph A. Ludwig; Eugenie S. Kleinerman; Shulin Li

To date, no specific marker exists for the detection of circulating tumor cells (CTC) from different types of sarcomas, though tools are available for detection of CTCs in peripheral blood of patients with cancer for epithelial cancers. Here, we report cell-surface vimentin (CSV) as an exclusive marker on sarcoma CTC regardless of the tissue origin of the sarcoma as detected by a novel monoclonal antibody. Utilizing CSV as a probe, we isolated and enumerated sarcoma CTCs with high sensitivity and specificity from the blood of patients bearing different types of sarcoma, validating their phenotype by single cell genomic amplification, mutation detection, and FISH. Our results establish the first universal and specific CTC marker described for enumerating CTCs from different types of sarcoma, thereby providing a key prognosis tool to monitor cancer metastasis and relapse.


Hepatology | 2012

Interleukin-30: A novel antiinflammatory cytokine candidate for prevention and treatment of inflammatory cytokine-induced liver injury

Denada Dibra; Jeffry Cutrera; Xueqing Xia; Bhaskar Kallakury; Lopa Mishra; Shulin Li

The liver is the major metabolic organ and is subjected to constant attacks from chronic viral infection, uptake of therapeutic drugs, life behavior (alcoholic), and environmental contaminants, all of which result in chronic inflammation, fibrosis, and, ultimately, cancer. Therefore, there is an urgent need to discover effective therapeutic agents for the prevention and treatment of liver injury, the ideal drug being a naturally occurring biological inhibitor. Here we establish the role of IL30 as a potent antiinflammatory cytokine that can inhibit inflammation‐induced liver injury. In contrast, interleukin (IL)27, which contains IL30 as a subunit, is not hepatoprotective. Interestingly, IL30 is induced by the proinflammatory signal such as IL12 through interferon‐gamma (IFN‐γ) / signal transducer and activator of transcription 1 signaling. In animal models, administration of IL30 by way of a gene therapy approach prevents and treats both IL12‐, IFN‐γ‐, and concanavalin A‐induced liver toxicity. Likewise, immunohistochemistry analysis of human tissue samples revealed that IL30 is highly expressed in hepatocytes, yet barely expressed in inflammation‐induced tissue such as fibrous/connective tissue. Conclusion: These novel observations reveal a novel role of IL30 as a therapeutic cytokine that suppresses proinflammatory cytokine‐associated liver toxicity. (Hepatology 2012)


Molecular Therapy | 2011

Discovery of a Linear Peptide for Improving Tumor Targeting of Gene Products and Treatment of Distal Tumors by IL-12 Gene Therapy

Jeffry Cutrera; Denada Dibra; Xueqing Xia; Azeem Hasan; Scott D. Reed; Shulin Li

Like many effective therapeutics, interleukin-12 (IL-12) therapy often causes side effects. Tumor targeted delivery may improve the efficacy and decrease the toxicity of systemic IL-12 treatments. In this study, a novel targeting approach was investigated. A secreted alkaline phosphatase (SEAP) reporter gene-based screening process was used to identify a mini-peptide which can be produced in vivo to target gene products to tumors. The coding region for the best peptide was inserted into an IL-12 gene to determine the antitumor efficacy. Affinity chromatography, mass spectrometry analysis, and binding studies were used to identify a receptor for this peptide. We discovered that the linear peptide VNTANST increased the tumor accumulation of the reporter gene products in five independent tumor models including one human xenogeneic model. The product from VNTANST-IL-12 fusion gene therapy increased accumulation of IL-12 in the tumor environment, and in three tumor models, VNTANST-IL-12 gene therapy inhibited distal tumor growth. In a spontaneous lung metastasis model, inhibition of metastatic tumor growth was improved compared to wild-type IL-12 gene therapy, and in a squamous cell carcinoma model, toxic liver lesions were reduced. The receptor for VNTANST was identified as vimentin. These results show the promise of using VNTANST to improve IL-12 treatments.


Journal of the National Cancer Institute | 2015

FGL2 as a Multimodality Regulator of Tumor-Mediated Immune Suppression and Therapeutic Target in Gliomas

Jun Yan; Ling Yuan Kong; Jiemiao Hu; Konrad Gabrusiewicz; Denada Dibra; Xueqing Xia; Amy B. Heimberger; Shulin Li

BACKGROUND Fibrinogen-like protein 2 (FGL2) may promote glioblastoma multiforme (GBM) cancer development by inducing multiple immune-suppression mechanisms. METHODS The biological significance of FGL2 expression was assessed using the The Cancer Genome Atlast (TCGA) glioma database and tumor lysates analysis. The therapeutic effects of an anti-Fgl2 antibody and the role of immune suppression regulation by Fgl2 were determined in immune-competent, NOD-scid IL2Rgammanull (NSG), and FcɣRIIB-/- mice (n = 3-18 per group). Data were analyzed with two-way analysis of variance, log-rank survival analysis, and Pearson correlation. All statistical tests were two-sided. RESULTS In low-grade gliomas, 72.5% of patients maintained two copies of the FGL2 gene, whereas 83.8% of GBM patients had gene amplification or copy gain. Patients with high levels of FGL2 mRNA in glioma tissues had a lower overall survival (P = .009). Protein levels of FGL2 in GBM lysates were higher relative to low-grade glioma lysates (11.48±5.75ng/mg vs 3.96±1.01ng/mg, P = .003). In GL261 mice treated with an anti-FGL2 antibody, median survival was 27 days compared with only 17 days for mice treated with an isotype control antibody (P = .01). The anti-FGL2 antibody treatment reduced CD39(+) Tregs, M2 macrophages, programmed cell death protein 1 (PD-1), and myeloid-derived suppressor cells (MDSCs). FGL2-induced increases in M2, CD39, and PD-1 were ablated in FcɣRIIB-/- mice. CONCLUSIONS FGL2 augments glioma immunosuppression by increasing the expression levels of PD-1 and CD39, expanding the frequency of tumor-supportive M2 macrophages via the FcγRIIB pathway, and enhancing the number of MDSCs and CD39(+) regulatory T cells. Collectively, these results show that FGL2 functions as a key immune-suppressive modulator and has potential as an immunotherapeutic target for treating GBM.


Journal of Immunology | 2012

Coordination between TLR9 Signaling in Macrophages and CD3 Signaling in T Cells Induces Robust Expression of IL-30

Denada Dibra; Jeffry Cutrera; Shulin Li

IL-30, the p28 subunit of IL-27, interacts with EBV-induced gene 3 to form IL-27, which modulates both proinflammatory and anti-inflammatory responses during autoimmune or infectious disease. It also acts as a natural antagonist of gp130, thereby attenuating the signals of other gp130-associated cytokines. IL-30 regulation via LPS has been reported by others, but the intercellular communication that induces IL-30 expression is unknown. In this study, we show that treatment with anti-CD3/CD28 Abs plus CpG oligodeoxynucleotides induces robust expression of IL-30, whereas either treatment alone induces only low expression of IL-30. This observation in vitro mirrors the murine model in which administration of CpG under inflammatory conditions in vivo induces IL-30 expression. This robust induction of IL-30 occurs through the coordination of helper CD4+ T cells and innate immune cells (e.g., macrophages) and, to a lesser degree, B cells via the CD40/CD154 signaling pathway. These findings reveal a previously unrecognized mechanism that integrates signaling pathways from T cells and macrophages at the cellular level to induce IL-30 expression.


Biochimica et Biophysica Acta | 2014

Molecular mechanisms of oncogene-induced inflammation and inflammation-sustained oncogene activation in gastrointestinal tumors: An underappreciated symbiotic relationship

Denada Dibra; Lopa Mishra; Shulin Li

Inflammation plays an integral part in tumor initiation. Specifically, patients with colitis, pancreatitis, or hepatitis have an increased susceptibility to cancer. The activation, mutation, and overexpression of oncogenes have been well documented in cell proliferation and transformation. Recently, oncogenes were found to also regulate the inflammatory milieu in tumors. Similarly, the inflammatory milieu can promote oncogene activation. In this review, we summarize advances of the symbiotic relationship oncogene activation and inflammation in gastrointestinal tumors such as colorectal, hepatic, and pancreatic tumors. NF-κB and STAT3 are the two most common pathways that are deregulated via these oncogenes. Understanding these interactions may yield effective therapeutic strategies for tumor prevention and treatment.


Cell Communication and Signaling | 2013

The cell-to-cell coordination between activated T cells and CpG-stimulated macrophages synergistically induce elevated levels of IL-10 via NF-κB1, STAT3, and CD40/CD154

Denada Dibra; Shulin Li

BackgroundStudies into the regulation of interleukin-10 (IL-10), have focused only on the molecular or single-cell level. The cues that induce IL-10 in the context of cell-to-cell communication are scarce. To fill this gap, this study elucidates the cell-to-cell interaction dependent regulation of IL-10.ResultsThe simultaneous activation of CD4+ T cells via CD3/CD28 and stimulation of macrophages via CpG and their intercellular communication with each other in the same microenvironment is necessary to induce a synergistic expression of IL-10. NF-κB1, ERK, and STAT3 are positive regulators of this cell-to-cell communication mediated molecular change of IL-10 induction. Strikingly, the activation of CD40/CD154 signaling is a negative regulator of IL-10 levels by CD3/CD28/CpG.ConclusionsThese findings are of prominence as CD3/CD28/CpG treatment can induce the anti-inflammatory cytokines IL-10 and IL-30, and the activation or inhibition of the CD40/CD154 acts as molecular rheostat of the expression of IL-10 or IL-30. More importantly, this not only serves as an example of IL-10 regulation at the cellular via coordination of two signals from two cell types, but these findings also lay the molecular and cellular groundwork for future studies to investigate how to manipulate IL-10 or IL-30 production during inflammation, cancer, or autoimmune diseases.


Hepatology | 2016

Mutant p53 in concert with an interleukin‐27 receptor alpha deficiency causes spontaneous liver inflammation, fibrosis, and steatosis in mice

Denada Dibra; Xueqing Xia; Abhisek Mitra; Jeffry Cutrera; Guillermina Lozano; Shulin Li

The cellular and molecular etiology of unresolved chronic liver inflammation remains obscure. Whereas mutant p53 has gain‐of‐function properties in tumors, the role of this protein in liver inflammation is unknown. Herein, mutant p53R172H is mechanistically linked to spontaneous and sustained liver inflammation and steatosis when combined with the absence of interleukin‐27 (IL27) signaling (IL27RA), resembling the phenotype observed in nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) patients. Indeed, these mice develop, with age, hepatocyte necrosis, immune cell infiltration, fibrosis, and micro‐ and macrosteatosis; however, these phenotypes are absent in mutant p53R172H or IL27RA‐/‐ mice. Mechanistically, endothelin A receptor (ETAR)‐positive macrophages are highly accumulated in the inflamed liver, and chemical inhibition of ETAR signaling reverses the observed phenotype and negatively regulates mutant p53 levels in macrophages. Conclusion: The combination of mutant p53 and IL27RA‐/‐ causes spontaneous liver inflammation, steatosis, and fibrosis in vivo, whereas either gene alone in vivo has no effects on the liver. (Hepatology 2016;63:1000–1012)


PLOS ONE | 2011

Wsx1 expression in tumors induces immune tolerance via suppression of effector immune cells

Denada Dibra; Jeffry Cutrera; Xueqing Xia; Shulin Li

Crosstalk between tumor cells and the cognate microenvironment plays a crucial role in tumor initiation and progression. However, only a few genes are known to affect such a crosstalk. This study reveals that WSX1 plays such a role when highly expressed in tumor cells. The expression of WSX1 in Lewis Lung Carcinoma (LLC) and the melanoma cell line AGS induces the death of T cells and inhibits the production of the effector cytokine IFNγ from NK and T cells, resulting in the promotion of tumor growth. These pro-tumorigenic properties of WSX1 are independent of IL27. This key observation reveals a new pathway of tumor-host interaction, which will ultimately lead to better strategies in immune therapy to reverse tumor tolerance.


Clinical Cancer Research | 2016

Lack of Immunomodulatory Interleukin-27 Enhances Oncogenic Properties of Mutant p53 In Vivo

Denada Dibra; Abhisek Mitra; Melissa Newman; Xueqing Xia; Jeffry Cutrera; Mihai Gagea; Eugenie S. Kleinerman; Guillermina Lozano; Shulin Li

Purpose: p53 is mutated in about 50% of human cancers, mostly through missense mutations. Expression of mutant p53 is associated with poor clinical outcomes or metastasis. Although mutant p53 is inherently instable, various stressors such as DNA damage or expression of the oncogenic Kras or c-myc affect the oncogenic properties of mutant p53. However, the effects of inflammation on mutant p53 are largely unknown. IL27 is an important immunomodulatory cytokine, but its impact on mutant p53-driven tumorigenesis has not been reported. Experimental Design: IL27RA−/− mice were bred with mutant p53 heterozygous (p53R172H/+) mice to obtain IL27RA−/−p53H/+ and IL27RA−/−p53H/H mice. Mouse survival and tumor spectra for the cohort were analyzed. Stability of p53 protein was analyzed via IHC and Western blot analysis. Results: This study unraveled that lack of IL27 signaling significantly shortened the survival duration of mice with tumors expressing both copies of the mutant p53 gene (Li-Fraumeni mouse model). Interestingly, in mice that were heterozygous for mutant p53, lack of IL27 signaling not only significantly shortened survival time but also doubled the incidence of osteosarcomas. Furthermore, lack of IL27 signaling is closely associated with increased mutant p53 stability in vivo from early age. Conclusions: These results suggest that IL27 signaling modulates the oncogenic properties of mutant p53 in vivo. Clin Cancer Res; 22(15); 3876–83. ©2016 AACR.

Collaboration


Dive into the Denada Dibra's collaboration.

Top Co-Authors

Avatar

Shulin Li

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Xueqing Xia

Louisiana State University

View shared research outputs
Top Co-Authors

Avatar

Jeffry Cutrera

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Abhisek Mitra

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Melissa Newman

Louisiana State University

View shared research outputs
Top Co-Authors

Avatar

Guillermina Lozano

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Amy B. Heimberger

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Arun Satelli

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Azeem Hasan

Louisiana State University

View shared research outputs
Top Co-Authors

Avatar

Camille Keenan

University of Texas at Austin

View shared research outputs
Researchain Logo
Decentralizing Knowledge