Network


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

Hotspot


Dive into the research topics where Biji Mathew is active.

Publication


Featured researches published by Biji Mathew.


Anesthesia & Analgesia | 2011

The novel role of the mu opioid receptor in lung cancer progression: a laboratory investigation.

Biji Mathew; Frances E. Lennon; Jessica Siegler; Tamara Mirzapoiazova; Nurbek Mambetsariev; Saad Sammani; Lynnette M. Gerhold; Patrick J. LaRiviere; Chin-Tu Chen; Joe G. N. Garcia; Ravi Salgia; Jonathan Moss; Patrick A. Singleton

BACKGROUND:The possibility that &mgr; opioid agonists can influence cancer recurrence is a subject of recent interest. Epidemiologic studies suggested that there were differences in cancer recurrence in breast and prostate cancer contingent on anesthetic regimens. In this study, we identify a possible mechanism for these epidemiologic findings on the basis of &mgr; opioid receptor (MOR) regulation of Lewis lung carcinoma (LLC) tumorigenicity in cell and animal models. METHODS:We used human lung tissue and human non–small cell lung cancer (NSCLC) cell lines and evaluated MOR expression using immunoblot and immunohistochemical analysis. LLC cells were treated with the peripheral opioid antagonist methylnaltrexone (MNTX) or MOR shRNA and evaluated for proliferation, invasion, and soft agar colony formation in vitro and primary tumor growth and lung metastasis in C57BL/6 and MOR knockout mice using VisEn fluorescence mediated tomography imaging and immunohistochemical analysis. RESULTS:We provide several lines of evidence that the MOR may be a potential target for lung cancer, a disease with high mortality and few treatment options. We first observed that there is ∼5- to 10-fold increase in MOR expression in lung samples from patients with NSCLC and in several human NSCLC cell lines. The MOR agonists morphine and [D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) increased in vitro LLC cell growth. Treatment with MNTX or silencing MOR expression inhibited LLC invasion and anchorage-independent growth by 50%–80%. Injection of MOR silenced LLC lead to a ∼65% reduction in mouse lung metastasis. In addition, MOR knockout mice do not develop significant tumors when injected with LLC in comparison with wild-type controls. Finally, continuous infusion of the peripheral opioid antagonist MNTX attenuates primary LLC tumor growth and reduces lung metastasis. CONCLUSIONS:Taken together, our data suggest a possible direct effect of opiates on lung cancer progression, and provide a plausible explanation for the epidemiologic findings. Our observations further suggest a possible therapeutic role for opioid antagonists.


American Journal of Respiratory Cell and Molecular Biology | 2010

Differential Effects of Sphingosine 1–Phosphate Receptors on Airway and Vascular Barrier Function in the Murine Lung

Saad Sammani; Liliana Moreno-Vinasco; Tamara Mirzapoiazova; Patrick A. Singleton; Eddie T. Chiang; Carrie Evenoski; Ting Wang; Biji Mathew; Aliya N. Husain; Jaideep Moitra; Xiaoguang Sun; Luis Nuñez; Jeffrey R. Jacobson; Steven M. Dudek; Viswanathan Natarajan; Joe G. N. Garcia

The therapeutic options for ameliorating the profound vascular permeability, alveolar flooding, and organ dysfunction that accompanies acute inflammatory lung injury (ALI) remain limited. Extending our previous finding that the intravenous administration of the sphingolipid angiogenic factor, sphingosine 1-phosphate (S1P), attenuates inflammatory lung injury and vascular permeability via ligation of S1PR(1), we determine that a direct intratracheal or intravenous administration of S1P, or a selective S1P receptor (S1PR(1)) agonist (SEW-2871), produces highly concentration-dependent barrier-regulatory responses in the murine lung. The intratracheal or intravenous administration of S1P or SEW-2871 at < 0.3 mg/kg was protective against LPS-induced murine lung inflammation and permeability. However, intratracheal delivery of S1P at 0.5 mg/kg (for 2 h) resulted in significant alveolar-capillary barrier disruption (with a 42% increase in bronchoalveolar lavage protein), and produced rapid lethality when delivered at 2 mg/kg. Despite the greater selectivity for S1PR(1), intratracheally delivered SEW-2871 at 0.5 mg/kg also resulted in significant alveolar-capillary barrier disruption, but was not lethal at 2 mg/kg. Consistent with the S1PR(1) regulation of alveolar/vascular barrier function, wild-type mice pretreated with the S1PR(1) inverse agonist, SB-649146, or S1PR(1)(+/-) mice exhibited reduced S1P/SEW-2871-mediated barrier protection after challenge with LPS. In contrast, S1PR(2)(-/-) knockout mice as well as mice with reduced S1PR(3) expression (via silencing S1PR3-containing nanocarriers) were protected against LPS-induced barrier disruption compared with control mice. These studies underscore the potential therapeutic effects of highly selective S1PR(1) receptor agonists in reducing inflammatory lung injury, and highlight the critical role of the S1P delivery route, S1PR(1) agonist concentration, and S1PR(1) expression in target tissues.


American Journal of Respiratory Cell and Molecular Biology | 2013

Sphingosine-1–Phosphate, FTY720, and Sphingosine-1–Phosphate Receptors in the Pathobiology of Acute Lung Injury

Viswanathan Natarajan; Steven M. Dudek; Jeffrey R. Jacobson; Liliana Moreno-Vinasco; Long Shuang Huang; Taimur Abassi; Biji Mathew; Yutong Zhao; Lichun Wang; Robert Bittman; Ralph R. Weichselbaum; Evgeny Berdyshev; Joe G. N. Garcia

Acute lung injury (ALI) attributable to sepsis or mechanical ventilation and subacute lung injury because of ionizing radiation (RILI) share profound increases in vascular permeability as a key element and a common pathway driving increased morbidity and mortality. Unfortunately, despite advances in the understanding of lung pathophysiology, specific therapies do not yet exist for the treatment of ALI or RILI, or for the alleviation of unremitting pulmonary leakage, which serves as a defining feature of the illness. A critical need exists for new mechanistic insights that can lead to novel strategies, biomarkers, and therapies to reduce lung injury. Sphingosine 1-phosphate (S1P) is a naturally occurring bioactive sphingolipid that acts extracellularly via its G protein-coupled S1P1-5 as well as intracellularly on various targets. S1P-mediated cellular responses are regulated by the synthesis of S1P, catalyzed by sphingosine kinases 1 and 2, and by the degradation of S1P mediated by lipid phosphate phosphatases, S1P phosphatases, and S1P lyase. We and others have demonstrated that S1P is a potent angiogenic factor that enhances lung endothelial cell integrity and an inhibitor of vascular permeability and alveolar flooding in preclinical animal models of ALI. In addition to S1P, S1P analogues such as 2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol (FTY720), FTY720 phosphate, and FTY720 phosphonates offer therapeutic potential in murine models of lung injury. This translational review summarizes the roles of S1P, S1P analogues, S1P-metabolizing enzymes, and S1P receptors in the pathophysiology of lung injury, with particular emphasis on the development of potential novel biomarkers and S1P-based therapies for ALI and RILI.


The FASEB Journal | 2013

Targeting sphingosine kinase 1 attenuates bleomycin-induced pulmonary fibrosis

Long Shuang Huang; Evgeny Berdyshev; Biji Mathew; Panfeng Fu; Irina Gorshkova; Donghong He; Wenli Ma; Imre Noth; Shwu Fan Ma; Srikanth Pendyala; Sekhar P. Reddy; Tong Zhou; Wei Zhang; Steven A. Garzon; Joe G. N. Garcia; Viswanathan Natarajan

Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive interstitial lung disease, wherein transforming growth factor β (TGF‐β) and sphingosine‐1‐phosphate (S1P) contribute to the pathogenesis of fibrosis. However, the in vivo contribution of sphingosine kinase (SphK) in fibrotic processes has not been documented. Microarray analysis of blood mononuclear cells from patients with IPF and SphK1‐ or SphK2‐knockdown mice and SphK inhibitor were used to assess the role of SphKs in fibrogenesis. The expression of SphK1/2 negatively correlated with lung function and survival in patients with IPF. Also, the expression of SphK1 was increased in lung tissues from patients with IPF and bleomycin‐challenged mice. Knockdown of SphK1, but not SphK2, increased survival and resistance to pulmonary fibrosis in bleomycin‐challenged mice. Administration of SphK inhibitor reduced bleomycin‐induced mortality and pulmonary fibrosis in mice. Knockdown of SphK1 or treatment with SphK inhibitor attenuated S1P generation and TGF‐β secretion in a bleomycin‐induced lung fibrosis mouse model that was accompanied by reduced phosphorylation of Smad2 and MAPKs in lung tissue. In vitro, bleomycin‐induced expression of SphK1 in lung fibroblast was found to be TGF‐β dependent. Taken together, these data indicate that SphK1 plays a critical role in the pathology of lung fibrosis and is a novel therapeutic target.—Huang, L. S., Berdyshev, E., Mathew, B., Fu, P., Gorshkova, I. A., He, D., Ma, W., Noth, I., Ma, S.‐F., Pendyala, S., Reddy, S. P., Zhou, T., Zhang, W., Garzon, S. A., Garcia, J. G. N., Natarajan, V. Targeting sphingosine kinase 1 attenuates bleomycin‐induced pulmonary fibrosis. FASEB J. 27, 1749–1760 (2013). www.fasebj.org


American Journal of Respiratory Cell and Molecular Biology | 2011

Simvastatin attenuates radiation-induced murine lung injury and dysregulated lung gene expression

Biji Mathew; Yong Huang; Jeffrey R. Jacobson; Evegeny Berdyshev; Lynnette M. Gerhold; Ting Wang; Liliana Moreno-Vinasco; Gabriel Lang; Yutong Zhao; Chin-Tu Chen; Patrick J. LaRiviere; Helena J. Mauceri; Saad Sammani; Aliya N. Husain; Steven M. Dudek; Viswanathan Natarajan; Yves A. Lussier; Ralph R. Weichselbaum; Joe G. N. Garcia

Novel therapies are desperately needed for radiation-induced lung injury (RILI), which, despite aggressive corticosteroid therapy, remains a potentially fatal and dose-limiting complication of thoracic radiotherapy. We assessed the utility of simvastatin, an anti-inflammatory and lung barrier-protective agent, in a dose- and time-dependent murine model of RILI (18-(25 Gy). Simvastatin reduced multiple RILI indices, including vascular leak, leukocyte infiltration, and histological evidence of oxidative stress, while reversing RILI-associated dysregulated gene expression, including p53, nuclear factor-erythroid-2-related factor, and sphingolipid metabolic pathway genes. To identify key regulators of simvastatin-mediated RILI protection, we integrated whole-lung gene expression data obtained from radiated and simvastatin-treated mice with protein-protein interaction network analysis (single-network analysis of proteins). Topological analysis of the gene product interaction network identified eight top-prioritized genes (Ccna2a, Cdc2, fcer1 g, Syk, Vav3, Mmp9, Itgam, Cd44) as regulatory nodes within an activated RILI network. These studies identify the involvement of specific genes and gene networks in RILI pathobiology, and confirm that statins represent a novel strategy to limit RILI.


The FASEB Journal | 2011

Role of sphingolipids in murine radiation-induced lung injury: protection by sphingosine 1-phosphate analogs

Biji Mathew; Jeffrey R. Jacobson; Evgeny Berdyshev; Yong Huang; Xiaoguang Sun; Yutong Zhao; Lynnette M. Gerhold; Jessica Siegler; Carrie Evenoski; Ting Wang; Tong Zhou; Rafe Zaidi; Liliana Moreno-Vinasco; Robert Bittman; Chin-Tu Chen; Patrick J. LaRiviere; Saad Sammani; Yves A. Lussier; Steven M. Dudek; Viswanathan Natarajan; Ralph R. Weichselbaum; Joe G. N. Garcia

Clinically significant radiation‐induced lung injury (RILI) is a common toxicity in patients administered thoracic radiotherapy. Although the molecular etiology is poorly understood, we previously characterized a murine model of RILI in which alterations in lung barrier integrity surfaced as a potentially important pathobiological event and genome‐wide lung gene mRNA levels identified dysregulation of sphingolipid metabolic pathway genes. We hypothesized that sphingolipid signaling components serve as modulators and novel therapeutic targets of RILI. Sphingolipid involvement in murine RILI was confirmed by radiation‐induced increases in lung expression of sphingosine kinase (SphK) isoforms 1 and 2 and increases in the ratio of ceramide to sphingosine 1‐phosphate (S1P) and dihydro‐S1P (DHS1P) levels in plasma, bronchoalveolar lavage fluid, and lung tissue. Mice with a targeted deletion of SphK1 (SphK1–/–) or with reduced expression of S1P receptors (S1PR1+/–, S1PR2–/–, and S1PR3–/–) exhibited marked RILI susceptibility. Finally, studies of 3 potent vascular barrier‐protective S1P analogs, FTY720, (S)‐FTY720‐phosphonate (fTyS), and SEW‐2871, identified significant RILI attenuation and radiation‐induced gene dysregulation by the phosphonate analog, fTyS (0.1 and 1 mg/kg i.p., 2×/wk) and to a lesser degree by SEW‐2871 (1 mg/kg i.p., 2×/wk), compared with those in controls. These results support the targeting of S1P signaling as a novel therapeutic strategy in RILI.—Mathew, B., Jacobson, J. R., Berdyshev, E., Huang, Y., Sun, X., Zhao, Y., Gerhold, L. M., Siegler, J., Evenoski, C., Wang, T., Zhou, T., Zaidi, R., Moreno‐Vinasco, L., Bittman, R., Chen, C. T., LaRiviere, P. J., Sammani, S., Lussier, Y. A., Dudek, S. M., Natarajan, V., Weichselbaum, R. R., Garcia, J. G. N. Role of sphingolipids in murine radiation‐induced lung injury: protection by sphingosine 1‐phosphate analogs. FASEB J. 25, 3388–3400 (2011). www.fasebj.org


Journal of Angiogenesis Research | 2010

Methylnaltrexone Potentiates the Anti-Angiogenic Effects of mTOR Inhibitors

Patrick A. Singleton; Nurbek Mambetsariev; Frances E. Lennon; Biji Mathew; Jessica Siegler; Liliana Moreno-Vinasco; Ravi Salgia; Jonathan Moss; Joe G. N. Garcia

BackgroundRecent cancer therapies include drugs that target both tumor growth and angiogenesis including mammalian target of rapamycin (mTOR) inhibitors. Since mTOR inhibitor therapy is associated with significant side effects, we examined potential agents that can reduce the therapeutic dose.MethodsMethylnaltrexone (MNTX), a peripheral mu opioid receptor (MOR) antagonist, in combination with the mTOR inhibitors temsirolimus and/or rapamycin, was evaluated for inhibition of VEGF-induced human pulmonary microvascular endothelial cell (EC) proliferation and migration as well as in vivo angiogenesis (mouse Matrigel plug assay).ResultsMNTX inhibited VEGF-induced EC proliferation and migration with an IC50 of ~100 nM. Adding 10 nM MNTX to EC shifted the IC50 of temsirolimus inhibition of VEGF-induced proliferation and migration from ~10 nM to ~1 nM and from ~50 to ~10 nM respectively. We observed similar effects with rapamycin. On a mechanistic level, we observed that MNTX increased EC plasma membrane-associated tyrosine phosphate activity. Inhibition of tyrosine phosphatase activity (3,4-dephostatin) blocked the synergy between MNTX and temsirolimus and increased VEGF-induced tyrosine phosphorylation of Src with enhanced PI3 kinase and mTOR Complex 2-dependent phosphorylation of Akt and subsequent activation of mTOR Complex 1 (rapamycin and temsirolimus target), while silencing Src, Akt or mTOR complex 2 components blocked VEGF-induced angiogenic events.ConclusionsOur data indicate that MNTX exerts a synergistic effect with rapamycin and temsirolimus on inhibition of VEGF-induced human EC proliferation and migration and in vivo angiogenesis. Therefore, addition of MNTX could potentially lower the dose of mTOR inhibitors which could improve therapeutic index.


Particle and Fibre Toxicology | 2012

Particulate matter air pollution disrupts endothelial cell barrier via calpain-mediated tight junction protein degradation

Ting Wang; Lichun Wang; Liliana Moreno-Vinasco; Gabriel Lang; Jessica Siegler; Biji Mathew; Peter V. Usatyuk; Jonathan M. Samet; Alison S. Geyh; Patrick N. Breysse; Viswanathan Natarajan; Joe G. N. Garcia

BackgroundExposure to particulate matter (PM) is a significant risk factor for increased cardiopulmonary morbidity and mortality. The mechanism of PM-mediated pathophysiology remains unknown. However, PM is proinflammatory to the endothelium and increases vascular permeability in vitro and in vivo via ROS generation.ObjectivesWe explored the role of tight junction proteins as targets for PM-induced loss of lung endothelial cell (EC) barrier integrity and enhanced cardiopulmonary dysfunction.MethodsChanges in human lung EC monolayer permeability were assessed by Transendothelial Electrical Resistance (TER) in response to PM challenge (collected from Ft. McHenry Tunnel, Baltimore, MD, particle size >0.1 μm). Biochemical assessment of ROS generation and Ca2+ mobilization were also measured.ResultsPM exposure induced tight junction protein Zona occludens-1 (ZO-1) relocation from the cell periphery, which was accompanied by significant reductions in ZO-1 protein levels but not in adherens junction proteins (VE-cadherin and β-catenin). N-acetyl-cysteine (NAC, 5 mM) reduced PM-induced ROS generation in ECs, which further prevented TER decreases and atteneuated ZO-1 degradation. PM also mediated intracellular calcium mobilization via the transient receptor potential cation channel M2 (TRPM2), in a ROS-dependent manner with subsequent activation of the Ca2+-dependent protease calpain. PM-activated calpain is responsible for ZO-1 degradation and EC barrier disruption. Overexpression of ZO-1 attenuated PM-induced endothelial barrier disruption and vascular hyperpermeability in vivo and in vitro.ConclusionsThese results demonstrate that PM induces marked increases in vascular permeability via ROS-mediated calcium leakage via activated TRPM2, and via ZO-1 degradation by activated calpain. These findings support a novel mechanism for PM-induced lung damage and adverse cardiovascular outcomes.


Journal of Lipid Research | 2012

Inhibition of serine palmitoyltransferase delays the onset of radiation-induced pulmonary fibrosis through the negative regulation of sphingosine kinase-1 expression

Irina Gorshkova; Tong Zhou; Biji Mathew; Jeffrey R. Jacobson; Daisuke Takekoshi; Palash Bhattacharya; B Smith; Bulent Aydogan; Ralph R. Weichselbaum; Viswanathan Natarajan; Joe G. N. Garcia; Evgeny Berdyshev

The enforcement of sphingosine-1-phosphate (S1P) signaling network protects from radiation-induced pneumonitis. We now demonstrate that, in contrast to early postirradiation period, late postirradiation sphingosine kinase-1 (SphK1) and sphingoid base-1-phosphates are associated with radiation-induced pulmonary fibrosis (RIF). Using the mouse model, we demonstrate that RIF is characterized by a marked upregulation of S1P and dihydrosphingosine-1-phosphate (DHS1P) levels in the lung tissue and in circulation accompanied by increased lung SphK1 expression and activity. Inhibition of sphingolipid de novo biosynthesis by targeting serine palmitoyltransferase (SPT) with myriocin reduced radiation-induced pulmonary inflammation and delayed the onset of RIF as evidenced by increased animal lifespan and decreased expression of markers of fibrogenesis, such as collagen and α-smooth muscle actin (α-SMA), in the lung. Long-term inhibition of SPT also decreased radiation-induced SphK activity in the lung and the levels of S1P-DHS1P in the lung tissue and in circulation. In vitro, inhibition or silencing of serine palmitoyltransferase attenuated transforming growth factor-β1 (TGF-β)-induced upregulation of α-SMA through the negative regulation of SphK1 expression in normal human lung fibroblasts. These data demonstrate a novel role for SPT in regulating TGF-β signaling and fibrogenesis that is linked to the regulation of SphK1 expression and S1P-DHS1P formation.


Thorax | 2015

Sphingosine-1-phosphate lyase is an endogenous suppressor of pulmonary fibrosis: role of S1P signalling and autophagy

Long Shuang Huang; Evgeny Berdyshev; John T. Tran; Lishi Xie; Jiwang Chen; David L. Ebenezer; Biji Mathew; Irina Gorshkova; Wei Zhang; Sekhar P. Reddy; Anantha Harijith; Gang Wang; Carol A. Feghali-Bostwick; Imre Noth; Shwu Fan Ma; Tong Zhou; Wenli Ma; Joe G. N. Garcia; Viswanathan Natarajan

Introduction Idiopathic pulmonary fibrosis (IPF) is characterised by accumulation of fibroblasts and myofibroblasts and deposition of extracellular matrix proteins. Sphingosine-1-phosphate (S1P) signalling plays a critical role in pulmonary fibrosis. Methods S1P lyase (S1PL) expression in peripheral blood mononuclear cells (PBMCs) was correlated with pulmonary functions and overall survival; used a murine model to check the role of S1PL on the fibrogenesis and a cell culture system to study the effect of S1PL expression on transforming growth factor (TGF)-β- and S1P-induced fibroblast differentiation. Results S1PL expression was upregulated in fibrotic lung tissues and primary lung fibroblasts isolated from patients with IPF and bleomycin-challenged mice. TGF-β increased the expression of S1PL in human lung fibroblasts via activation and binding of Smad3 transcription factor to Sgpl1 promoter. Overexpression of S1PL attenuated TGF-β-induced and S1P-induced differentiation of human lung fibroblasts through regulation of the expression of LC3 and beclin 1. Knockdown of S1PL (Sgpl1+/−) in mice augmented bleomycin-induced pulmonary fibrosis, and patients with IPF reduced Sgpl1 mRNA expression in PBMCs exhibited higher severity of fibrosis and lower survival rate. Conclusion These studies suggest that S1PL is a novel endogenous suppressor of pulmonary fibrosis in human IPF and animal models.

Collaboration


Dive into the Biji Mathew's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Liliana Moreno-Vinasco

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Viswanathan Natarajan

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Jeffrey R. Jacobson

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Saad Sammani

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Ting Wang

University of Arizona

View shared research outputs
Top Co-Authors

Avatar

Jessica Siegler

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steven M. Dudek

University of Illinois at Chicago

View shared research outputs
Researchain Logo
Decentralizing Knowledge