Network


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

Hotspot


Dive into the research topics where Calvin R. Justus is active.

Publication


Featured researches published by Calvin R. Justus.


Frontiers in Physiology | 2013

Acidic tumor microenvironment and pH-sensing G protein-coupled receptors.

Calvin R. Justus; Lixue Dong; Li V. Yang

The tumor microenvironment is acidic due to glycolytic cancer cell metabolism, hypoxia, and deficient blood perfusion. It is proposed that acidosis in the tumor microenvironment is an important stress factor and selection force for cancer cell somatic evolution. Acidic pH has pleiotropic effects on the proliferation, migration, invasion, metastasis, and therapeutic response of cancer cells and the function of immune cells, vascular cells, and other stromal cells. However, the molecular mechanisms by which cancer cells and stromal cells sense and respond to acidic pH in the tumor microenvironment are poorly understood. In this article the role of a family of pH-sensing G protein-coupled receptors (GPCRs) in tumor biology is reviewed. Recent studies show that the pH-sensing GPCRs, including GPR4, GPR65 (TDAG8), GPR68 (OGR1), and GPR132 (G2A), regulate cancer cell metastasis and proliferation, immune cell function, inflammation, and blood vessel formation. Activation of the proton-sensing GPCRs by acidosis transduces multiple downstream G protein signaling pathways. Since GPCRs are major drug targets, small molecule modulators of the pH-sensing GPCRs are being actively developed and evaluated. Research on the pH-sensing GPCRs will continue to provide important insights into the molecular interaction between tumor and its acidic microenvironment and may identify new targets for cancer therapy and chemoprevention.


Journal of Visualized Experiments | 2014

In vitro cell migration and invasion assays.

Calvin R. Justus; Nancy R. Leffler; Maria J. Ruiz-Echevarria; Li V. Yang

Migration is a key property of live cells and critical for normal development, immune response, and disease processes such as cancer metastasis and inflammation. Methods to examine cell migration are very useful and important for a wide range of biomedical research such as cancer biology, immunology, vascular biology, cell biology and developmental biology. Here we use tumor cell migration and invasion as an example and describe two related assays to illustrate the commonly used, easily accessible methods to measure these processes. The first method is the cell culture wound closure assay in which a scratch is generated on a confluent cell monolayer. The speed of wound closure and cell migration can be quantified by taking snapshot pictures with a regular inverted microscope at several time intervals. More detailed cell migratory behavior can be documented using the time-lapse microscopy system. The second method described in this paper is the transwell cell migration and invasion assay that measures the capacity of cell motility and invasiveness toward a chemo-attractant gradient. It is our goal to describe these methods in a highly accessible manner so that the procedures can be successfully performed in research laboratories even just with basic cell biology setup.


International Journal of Molecular Sciences | 2015

Molecular Connections between Cancer Cell Metabolism and the Tumor Microenvironment

Calvin R. Justus; Edward J. Sanderlin; Li V. Yang

Cancer cells preferentially utilize glycolysis, instead of oxidative phosphorylation, for metabolism even in the presence of oxygen. This phenomenon of aerobic glycolysis, referred to as the “Warburg effect”, commonly exists in a variety of tumors. Recent studies further demonstrate that both genetic factors such as oncogenes and tumor suppressors and microenvironmental factors such as spatial hypoxia and acidosis can regulate the glycolytic metabolism of cancer cells. Reciprocally, altered cancer cell metabolism can modulate the tumor microenvironment which plays important roles in cancer cell somatic evolution, metastasis, and therapeutic response. In this article, we review the progression of current understandings on the molecular interaction between cancer cell metabolism and the tumor microenvironment. In addition, we discuss the implications of these interactions in cancer therapy and chemoprevention.


Biochimica et Biophysica Acta | 2017

GPR4 deficiency alleviates intestinal inflammation in a mouse model of acute experimental colitis

Edward J. Sanderlin; Nancy R. Leffler; Kvin Lertpiriyapong; Qi Cai; Heng Hong; Vasudevan Bakthavatchalu; James G. Fox; Joani Zary Oswald; Calvin R. Justus; Elizabeth A. Krewson; Dorcas P. O’Rourke; Li V. Yang

GPR4 is a proton-sensing G protein-coupled receptor that can be activated by extracellular acidosis. It has recently been demonstrated that activation of GPR4 by acidosis increases the expression of numerous inflammatory and stress response genes in vascular endothelial cells (ECs) and also augments EC-leukocyte adhesion. Inhibition of GPR4 by siRNA or small molecule inhibitors reduces endothelial cell inflammation. As acidotic tissue microenvironments exist in many types of inflammatory disorders, including inflammatory bowel disease (IBD), we examined the role of GPR4 in intestinal inflammation using a dextran sulfate sodium (DSS)-induced acute colitis mouse model. We observed that GPR4 mRNA expression was increased in mouse and human IBD tissues when compared to control intestinal tissues. To determine the function of GPR4 in intestinal inflammation, wild-type and GPR4-deficient mice were treated with 3% DSS for 7days to induce acute colitis. Our results showed that the severity of colitis was decreased in GPR4-deficient DSS-treated mice in comparison to wild-type DSS-treated mice. Clinical parameters, macroscopic disease indicators, and histopathological features were less severe in the DSS-treated GPR4-deficient mice than the DSS-treated wild-type mice. Endothelial adhesion molecule expression, leukocyte infiltration, and isolated lymphoid follicle (ILF) formation were reduced in intestinal tissues of DSS-treated GPR4-null mice. Collectively, our results suggest GPR4 provides a pro-inflammatory role in the inflamed gut as the absence of GPR4 ameliorates intestinal inflammation in the acute experimental colitis mouse model.


Experimental Cell Research | 2015

GPR4 decreases B16F10 melanoma cell spreading and regulates focal adhesion dynamics through the G13/Rho signaling pathway.

Calvin R. Justus; Li V. Yang

The effect of acidosis, a biochemical hallmark of the tumor microenvironment, on cancer progression and metastasis is complex. Both pro- and anti-tumorigenic effects of acidosis have been reported and the acidic microenvironment has been exploited for specific delivery of drugs, imaging agents, and genetic constructs into tumors. In this study we investigate the spreading and focal adhesion of B16F10 melanoma cells that are genetically engineered to overexpress the pH-sensing G protein-coupled receptor GPR4. By using cell attachment assays we found that GPR4 overexpression delayed cell spreading and altered the spatial localization of dynamic focal adhesion complex, such as the localization of phosphorylated focal adhesion kinase (FAK) and paxillin, at acidic pH. The potential G-protein and downstream signaling pathways that are responsible for these effects were also investigated. By using the Rho inhibitor CT04 (C3 transferase), the Rho-associated kinase (ROCK) inhibitors Y27632 and thiazovivin, the myosin light chain kinase (MLCK) inhibitor staurosporine or a G12/13 inhibitory construct, cell spreading was restored whereas the inhibition and activation of the Gq and Gs pathways had little or no effect. Altogether our results indicate that through the G12/13/Rho signaling pathway GPR4 modulates focal adhesion dynamics and reduces cell spreading and membrane ruffling.


Cell Health and Cytoskeleton | 2015

Emerging roles for the pH-sensing G protein-coupled receptors in response to acidotic stress

Edward J. Sanderlin; Calvin R. Justus; Elizabeth A. Krewson; Li V. Yang

License. The full terms of the License are available at http://creativecommons.org/licenses/by-nc/3.0/. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. Permissions beyond the scope of the License are administered by Dove Medical Press Limited. Information on how to request permission may be found at: http://www.dovepress.com/permissions.php Cell Health and Cytoskeleton 2015:7 99–109 Cell Health and Cytoskeleton Dovepress


Journal of Translational Medicine | 2017

Contextual tumor suppressor function of T cell death-associated gene 8 (TDAG8) in hematological malignancies

Calvin R. Justus; Edward J. Sanderlin; Lixue Dong; Tianai Sun; Jen-Tsan Chi; Kvin Lertpiriyapong; Li V. Yang

BackgroundExtracellular acidosis is a condition found within the tumor microenvironment due to inadequate blood perfusion, hypoxia, and altered tumor cell metabolism. Acidosis has pleiotropic effects on malignant progression; therefore it is essential to understand how acidosis exerts its diverse effects. TDAG8 is a proton-sensing G-protein-coupled receptor that can be activated by extracellular acidosis.MethodsTDAG8 gene expression was analyzed by bioinformatic analyses and quantitative RT-PCR in human hematological malignancies. Retroviral transduction was used to restore TDAG8 expression in U937, Ramos and other blood cancer cells. Multiple in vitro and in vivo tumorigenesis and metastasis assays were employed to evaluate the effects of TDAG8 expression on blood cancer progression. Western blotting, immunohistochemistry and biochemical approaches were applied to elucidate the underlying mechanisms associated with the TDAG8 receptor pathway.ResultsTDAG8 expression is significantly reduced in human blood cancers in comparison to normal blood cells. Severe acidosis, pH 6.4, inhibited U937 cancer cell proliferation while mild acidosis, pH 6.9, stimulated its proliferation. However, restoring TDAG8 gene expression modulated the U937 cell response to mild extracellular acidosis and physiological pH by reducing cell proliferation. Tumor xenograft experiments further revealed that restoring TDAG8 expression in U937 and Ramos cancer cells reduced tumor growth. It was also shown U937 cells with restored TDAG8 expression attached less to Matrigel, migrated slower toward a chemoattractant, and metastasized less in severe combined immunodeficient mice. These effects correlated with a reduction in c-myc oncogene expression. The mechanistic investigation indicated that Gα13/Rho signaling arbitrated the TDAG8-mediated c-myc oncogene repression in response to acidosis.ConclusionsThis study provides data to support the concept that TDAG8 functions as a contextual tumor suppressor down-regulated in hematological malignancies and potentiation of the TDAG8 receptor pathway may be explored as a potential anti-tumorigenic approach in blood cancers.


Cancer Research | 2017

Abstract 5916: Proton-sensor GPR4 potentiates intestinal inflammation in the DSS-induced colitis mouse model

Edward J. Sanderlin; Nancy R. Leffler; Kvin Lertpiriyapong; Qi Cai; Heng Hong; Vasudevan Bakthavatchalu; James G. Fox; Joani Zary Oswald; Calvin R. Justus; Elizabeth A. Krewson; Dorcas P. O’Rourke; Li V. Yang

Inflammation and tissue acidosis are two factors that co-exist in inflammatory bowel disease (IBD) and can contribute to increased risk of colorectal cancer (CRC) development. GPR4 is a proton-sensing G protein-coupled receptor that can be activated by extracellular acidosis through several histidine residues and subsequently signal through downstream G-protein pathways. Recently, GPR4 has been shown to be activated by acidosis and can increase the expression of numerous inflammatory and stress response genes in vascular endothelial cells (ECs) and has functionally increased EC-leukocyte adhesion. Subsequently, genetic and small molecule approaches for the inhibition of GPR4 activity have reduced endothelial cell inflammation. In this study, we examined the role of GPR4 in intestinal inflammation using a dextran sulfate sodium (DSS)-induced colitis mouse model. We observed that GPR4 mRNA expression was increased in mouse and human IBD tissues when compared to control intestinal tissues. To determine the function of GPR4 in intestinal inflammation, wild-type and GPR4-deficient mice were treated with 3% DSS for acute and chronic time points for the induction of colitis. Our results showed that the severity of colitis was decreased in GPR4-deficient DSS-treated mice in comparison to wild-type DSS-treated mice. Clinical parameters, macroscopic disease indicators, and histopathological features were less severe in the DSS-treated GPR4-deficient mice than the DSS-treated wild-type mice. Inflammatory gene expression, endothelial adhesion molecule expression, leukocyte infiltration, and isolated lymphoid follicle (ILF) formation were reduced in intestinal tissues of DSS-treated GPR4-null mice. In summary, our results suggest GPR4 potentiates intestinal inflammation as the absence of GPR4 ameliorates intestinal inflammation in the DSS-induced colitis mouse model. Use of GPR4 inhibitors could prove a valuable therapeutic in the reduction of intestinal inflammation and subsequent CRC development. Note: This abstract was not presented at the meeting. Citation Format: Edward J. Sanderlin, Nancy R. Leffler, Kvin Lertpiriyapong, Qi Cai, Heng Hong, Vasudevan Bakthavatchalu, James G. Fox, Joani Z. Oswald, Calvin R. Justus, Elizabeth A. Krewson, Dorcas O’Rourke, Li V. Yang. Proton-sensor GPR4 potentiates intestinal inflammation in the DSS-induced colitis mouse model [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5916. doi:10.1158/1538-7445.AM2017-5916


bioRxiv | 2016

GPR4 Deficiency Alleviates Intestinal Inflammation in a Mouse Model of Inflammatory Bowel Disease

Edward J. Sanderlin; Nancy R. Leffler; Kvin Lertpiriyapong; Qi Cai; Heng Hong; Vasudevan Bakthavatchalu; James G. Fox; Joani Zary Oswald; Calvin R. Justus; Elizabeth A. Krewson; D. O'Rourke; Li V. Yang

GPR4 is a proton-sensing G protein-coupled receptor that can be activated by extracellular acidosis. It has recently been demonstrated that activation of GPR4 by acidosis increases the expression of numerous inflammatory and stress response genes in vascular endothelial cells (ECs) and also augments EC-leukocyte adhesion. Inhibition of GPR4 by siRNA or small molecule inhibitors reduces endothelial cell inflammation. As acidotic tissue microenvironments exist in many types of inflammatory disorders, including inflammatory bowel disease (IBD), we examined the role of GPR4 in IBD using a dextran sulfate sodium (DSS)-induced colitis mouse model. We observed that GPR4 mRNA expression was increased in mouse and human IBD tissues when compared to control intestinal tissues. To determine the function of GPR4 in IBD, wild-type and GPR4-deficient mice were treated with 3% DSS for 7 days to induce acute colitis. Our results showed that the severity of colitis was decreased in GPR4-deficient DSS-treated mice in comparison to wild-type DSS-treated mice. Clinical parameters, macroscopic disease indicators, and histopathological features were less severe in the DSS-treated GPR4-deficient mice than the DSS-treated wild-type mice. Inflammatory gene expression, leukocyte infiltration, and isolated lymphoid follicle (ILF) formation were reduced in intestinal tissues of DSS-treated GPR4-null mice. Collectively, our results suggest GPR4 provides a pro-inflammatory role in IBD as the absence of GPR4 ameliorates intestinal inflammation in the acute DSS-induced IBD mouse model.


Cancer Research | 2013

Abstract 2799: Regulation of tumor cell attachment, spreading and migration by the GPR4 receptor and related G protein pathways.

Calvin R. Justus; Li V. Yang

The tumor microenvironment has a characteristic low pH due to inadequate blood perfusion, hypoxia, altered cellular metabolism, and inflammation. The effects of acidic pH on cancer progression are complex. It has been shown that low pH increases the metastatic spread of cancer by assisting in the degradation of extracellular matrix. Other studies show that acidic microenvironments decrease metastatic potential in B16F10 melanoma cells upon activation of the GPR4 receptor and also inhibit Akt activity in breast cancer cells. In this study we have investigated the attachment, spreading, and migration of B16F10 cells that have been genetically modified to express the GPR4 receptor at a high level (named B16F10/GPR4 cells). The results showed that upon stimulation of GPR4 in B16F10/GPR4 cells with low pH, cell spreading, membrane ruffling, and migration were significantly decreased. The phosphorylation levels of paxillin Y118 and focal adhesion kinase (FAK) Y397 following one-hour cell attachment were also decreased, and the spatial localization of the phosphorylated paxillin and FAK was substantially altered. Further tests were performed to identify the G-protein signaling pathway responsible for this effect. By using a G13 dominant negative construct or inhibiting Rho activation with C3 transferase (CT04) in B16F10/GPR4 cells, the cell spreading and membrane ruffling abilities were almost completely restored back to the level of B16F10/vector control cells, whereas the inhibition of the Gs and Gq pathways had little effect. These results suggest that activation of the GPR4 receptor and subsequently the G13/Rho downstream pathway are responsible for decreased cell attachment, spreading and membrane ruffling, which may cause decreased metastatic potential of B16F10/GPR4 cells found previously. Citation Format: Calvin R. Justus, Li V. Yang. Regulation of tumor cell attachment, spreading and migration by the GPR4 receptor and related G protein pathways. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2799. doi:10.1158/1538-7445.AM2013-2799

Collaboration


Dive into the Calvin R. Justus's collaboration.

Top Co-Authors

Avatar

Li V. Yang

East Carolina University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heng Hong

East Carolina University

View shared research outputs
Top Co-Authors

Avatar

James G. Fox

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qi Cai

East Carolina University

View shared research outputs
Top Co-Authors

Avatar

Vasudevan Bakthavatchalu

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge