Network


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

Hotspot


Dive into the research topics where Robert B. Crawford is active.

Publication


Featured researches published by Robert B. Crawford.


Journal of Pharmacology and Experimental Therapeutics | 2008

2,3,7,8-Tetrachlorodibenzo-p-dioxin-Mediated Impairment of B Cell Differentiation Involves Dysregulation of Paired Box 5 (Pax5) Isoform, Pax5a

Dina Schneider; Maria Alejandra Manzan; Robert B. Crawford; Weimin Chen; Norbert E. Kaminski

The persistent environmental contaminant and immunotoxicant, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), markedly suppresses humoral immune responses. We recently reported impaired down-regulation of paired box 5 (Pax5), a repressor of B cell differentiation and concomitant suppression of the IgM response by TCDD in the murine CH12.LX B cell line. The objectives of the current study were to determine the impact of TCDD treatment on molecular outcomes characteristic of terminal B cell differentiation and to assess the role that Pax5 isoforms plays in the suppression of B cell differentiation by TCDD. In this study, we show that the highly abundant full-length Pax5 isoform, Pax5a, and at least two additional modestly expressed Pax5 isoforms were expressed in CH12.LX and splenic B cells. In lipopolysaccharide (LPS)-activated B cells, all of the identified Pax5 isoforms were synchronously down-regulated, and in the presence of TCDD cotreatment they were abnormally and synchronously elevated, suggesting a common mechanism of regulation. Furthermore, B cell differentiation markers X-box protein-1 and major histocompatibility complex class II showed that the levels to which Pax5 was derepressed by TCDD were sufficient to impair B cell differentiation and immunoglobulin gene expression. Confirming the involvement of Pax5, ectopic expression of Pax5a in the LPS-activated CH12.LX cells closely mimicked the suppression of the IgM response by TCDD. In summary, our results demonstrate that Pax5a has a critical role in both the TCDD-mediated impairment of B cell differentiation and the suppression of the humoral immune response.


Journal of Leukocyte Biology | 2008

Effects of targeted deletion of cannabinoid receptors CB1 and CB2 on immune competence and sensitivity to immune modulation by Δ9‐tetrahydrocannabinol

Alison E.B. Springs; Peer W. F. Karmaus; Robert B. Crawford; Barbara L. F. Kaplan; Norbert E. Kaminski

The role of cannabinoid receptors, CB1 and CB2, in immune competence and modulation by Δ9‐tetrahydrocannabinol (Δ9‐THC) was investigated in CB1−/−/CB2−/− mice. Immunofluorescence analysis of splenic leukocytes showed no significant differences in the percentage of T cell subsets, B cells, or macrophages between wild‐type and CB1−/−/CB2−/− mice. Lymphoproliferative control responses to PHA, phorbol ester plus ionomycin, or LPS and sensitivity to suppression by Δ9‐THC showed no profound differences between the two genotypes, although some differences were observed in control baseline responses. Likewise, similar control responses and sensitivity to Δ9‐THC were observed in mixed lymphocyte responses (MLR) and in IL‐2 and IFN‐γ production in both genotypes. Conversely, humoral immune responses showed a markedly different profile of activity. Δ9‐THC suppressed the in vivo T cell‐dependent, anti‐sheep RBC (anti‐sRBC) IgM antibody‐forming cell (AFC) response in wild‐type but not in CB1−/−/CB2−/− mice, and the in vitro anti‐sRBC IgM response in CB1−/−/CB2−/− splenocytes was too low to rigorously assess CB1/CB2 involvement in modulation by Δ9‐THC. Conversely, comparable in vitro IgM AFC control responses to LPS and CD40 ligand (CD40L) activation were observed in the two genotypes. Interestingly, LPS‐induced IgM responses were refractory to suppression by Δ9‐THC, regardless of genotype, and CD40L‐induced IgM responses were only suppressed by Δ9‐THC in wild‐type but not in CB1−/−/CB2−/− B cells. Collectively, we demonstrate differential involvement of CB1 and/or CB2 in immune modulation by Δ9‐THC and in some control responses. Moreover, CB1/CB2 involvement was observed in humoral responses requiring CD40‐initiated signaling for suppression by Δ9‐THC.


Biochemical Pharmacology | 1996

Identification of functional aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator in murine splenocytes

Courtney E. Williams; Robert B. Crawford; Michael P. Holsapple; Norbert E. Kaminski

The objective of the present studies was to determine whether the aryl hydrocarbon receptor (AhR) and AhR nuclear translocator (ARNT) protein are present and functional in B6C3F1 (C57BL/6 x C3H) mouse splenocytes. Northern analysis of poly(A) RNA isolated from splenocytes revealed transcripts of approximately 6.6 kb which hybridized to the AhR complementary DNA (cDNA) probe. Anti-AhR antibodies identified two major cytosolic forms of the AhR in splenocytes, approximately 95 and 104 kDa, corresponding to the codominately expressed Ahrb alleles in the B6C3F1 mice. Northern analysis utilizing an oligomer probe for ARNT identified three messenger RNA (mRNA) transcripts, approximately 5.6, 2.0, and 1.1 kb, in spleen which was consistent with the banding pattern observed in the B6C3F1 mouse liver. Western blotting confirmed the presence of the approximately 87 kDa ARNT protein in splenocytes. Protein quantitation by slot blot analysis demonstrated approximately 2.0-fold more AhR in liver than in splenocytes. Interestingly, ARNT was approximately 2.4-fold more abundant in splenocytes than in liver. Consistent with these results, comparison by quantitative reverse transcriptase-polymerase chain reaction analysis of AhR and ARNT transcripts in liver and splenocytes demonstrated approximately 2.3-fold more AhR transcripts in liver than in splenocytes and approximately 3.2-fold more ARNT transcripts in splenocytes than in liver. In addition, comparisons between AhR and ARNT transcripts isolated from the liver and splenocytes indicated a greater number of ARNT transcripts as compared with AhR in both preparations. TCDD treatment of splenocytes induced binding of the AhR nuclear complex to the dioxin-responsive enhancer (DRE) as detected by the electrophoretic mobility shift assay. These findings confirm that the AhR and ARNT are present in mouse splenocytes and are capable of binding to the DRE.


Toxicology and Applied Pharmacology | 2011

The aryl hydrocarbon receptor interacts with ATP5α1, a subunit of the ATP synthase complex, and modulates mitochondrial function

Dorothy M. Tappenden; Scott G. Lynn; Robert B. Crawford; KangAe Lee; Ajith Vengellur; Norbert E. Kaminski; Russell S. Thomas; John J. LaPres

Dioxins, including 2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD), produce a wide range of toxic effects in mammals. Most, if not all, of these toxic effects are regulated by the aryl hydrocarbon receptor (AHR). The AHR is a ligand activated transcription factor that has been shown to interact with numerous proteins capable of influencing the receptors function. The ability of secondary proteins to alter AHR-mediated transcriptional events, a necessary step for toxicity, led us to determine whether additional interacting proteins could be identified. To this end, we have employed tandem affinity purification (TAP) of the AHR in Hepa1c1c7 cells. TAP of the AHR, followed by mass spectrometry (MS) identified ATP5α1, a subunit of the ATP synthase complex, as a strong AHR interactor in the absence of ligand. The interaction was lost upon exposure to TCDD. The association was confirmed by co-immunoprecipitation in multiple cell lines. In addition, cell fractionation experiments showed that a fraction of the AHR is found in the mitochondria. To ascribe a potential functional role to the AHR:ATP5α1 interaction, TCDD was shown to induce a hyperpolarization of the mitochondrial membrane in an AHR-dependent and transcription-independent manner. These results suggest that a fraction of the total cellular AHR pool is localized to the mitochondria and contributes to the organelles homeostasis.


International Journal of Cancer | 2003

Cannabinoids inhibit gap junctional intercellular communication and activate ERK in a rat liver epithelial cell line

Brad L. Upham; Alisa M. Rummel; Joseph M. Carbone; James E. Trosko; Yanli Ouyang; Robert B. Crawford; Norbert E. Kaminski

Many tumor promoters suppress the immune system; however, the direct effect of immunosuppressants on the tumorigenic pathways of nonimmune cells in solid tissue has not been well documented. Cannabinoids were chosen to explore this question further. Cannabinoids are immune modulators that affect specific intracellular signaling pathways in leukocytes. Since these compounds are nongenotoxic, any tumorigenic effect that might be associated with these compounds would need to occur through an epigenetic mechanism. Therefore, we determined the effect of Δ9‐THC and CBN, 2 plant‐derived cannabinoids, on 2 key epigenetic markers of tumor promotion: inhibition of GJIC, which is essential in removing a cell from growth suppression, and activation of the ERK–MAPK pathway, which is crucial in activating the appropriate genes for mitogenesis. Both Δ9‐THC and CBN reversibly inhibited GJIC at noncytotoxic doses (15 μM) in a normal diploid WB rat liver epithelial oval cell line within 20 min and activated ERK1 and ERK2 within 5 min. Inhibition of MEK with PD98059 prevented the inhibition of GJIC by either cannabinoid, suggesting that inhibition of GJIC was MEK‐dependent. Based on RT‐PCR analysis and employment of an antagonist of CB1 and CB2, the effects on GJIC and MAPK were independent of both cannabinoid receptors. Cannabinoids affected crucial epigenetic pathways associated with cell proliferation in a rodent liver epithelial cell model system.


Journal of Leukocyte Biology | 2011

Deletion of cannabinoid receptors 1 and 2 exacerbates APC function to increase inflammation and cellular immunity during influenza infection

Peer W. F. Karmaus; Weimin Chen; Robert B. Crawford; Jack R. Harkema; Barbara L. F. Kaplan; Norbert E. Kaminski

We and others have reported that simultaneous targeted deletion of CB1 and CB2 resulted in exacerbation of immune reactivity, suggesting a role of endocannabinoids in down‐regulating immune function. In this study, we demonstrate that APC function is enhanced specifically in the absence of CB1 and CB2 signaling, resulting in an exacerbated immune response phenotype. After influenza infection, CB1−/−CB2−/− mice showed more pronounced pulmonary damage, increased inflammatory cell infiltrate, inflammation, and a greater cellular immune responses compared with WT mice, as evidenced by transcriptome analysis, more robust T cell activation, and effector cell cytokine production. After direct activation in vitro, there were no differences in the percentages of cytokine‐producing CD4+ T cells between CB1−/−CB2−/− and WT mice. However, untreated CB1−/−CB2−/− mice routinely had fewer naïve T cells compared with WT, suggesting dysregulation of APC immune homeostasis. Moreover, bmDCs and AM isolated from CB1−/−CB2−/− mice exhibited a more mature phenotype, with and without TLR stimulation, and bmDCs elicited T cells more robustly than WT mice. Collectively, these findings implicate a role for CB1 and CB2 on APCs in regulating immune responses and immune homeostasis.


Toxicological Sciences | 2010

Induction of the Aryl Hydrocarbon Receptor-Responsive Genes and Modulation of the Immunoglobulin M Response by 2, 3, 7, 8-Tetrachlorodibenzo-p-Dioxin in Primary Human B Cells

Haitian Lu; Robert B. Crawford; Jose E. Suarez-Martinez; Barbara L. F. Kaplan; Norbert E. Kaminski

Past studies in rodent models identified the suppression of primary humoral immune responses as one of the most sensitive sequela associated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure. Yet, the sensitivity of humoral immunity to TCDD in humans represents an important toxicological data gap. Therefore, the objectives of this investigation were two-fold. The first was to assess the induction of known aryl hydrocarbon receptor (AHR)-responsive genes in primary human B cells as a measure of early biological responses to TCDD. The second was to evaluate the direct effect of TCDD on CD40 ligand-induced immunoglobulin M (IgM) secretion by human primary B cells. The effects of TCDD on induction of AHR-responsive genes and suppression of the IgM response were also compared with B cells from a TCDD-responsive mouse strain, C57BL/6. AHR-responsive genes in human B cells exhibited slower kinetics and reduced magnitude of induction by TCDD when compared with mouse B cells. Evaluation of B-cell function from 12 donors identified two general phenotypes; the majority of donors exhibited similar sensitivity to suppression by TCDD of the IgM response as mouse B cells, which was not attributable to decreased B-cell proliferation. In a minority of donors, no suppression of the IgM response by TCDD was observed. Although donor-to-donor variation in sensitivity to TCDD was observed, human B cells from the majority of donors evaluated showed impairment of effector function by TCDD. Collectively, data presented in this series of studies demonstrate that TCDD impairs the humoral immunity of humans by directly targeting B cells.


Toxicological Sciences | 2009

Involvement of Blimp-1 and AP-1 dysregulation in the 2,3,7,8-tetrachlorodibenzo-p-dioxin-mediated suppression of the IgM response by B cells

Dina Schneider; Maria Alejandra Manzan; Byung Sun Yoo; Robert B. Crawford; Norbert E. Kaminski

B cell differentiation and humoral immune responses are markedly suppressed by the persistent environmental contaminant, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). The suppression of humoral immune responses by TCDD occurs by direct actions on the B cell and involves activation of the aryl hydrocarbon receptor. Transcriptional regulation of paired box gene 5 (Pax5), an important regulator of B cell differentiation, is altered by TCDD in concordance with the suppression of B cell differentiation and humoral immunoglobulin M response. We hypothesized that TCDD treatment leads to dysregulation of Pax5 transcription by interfering with the basic B cell differentiation mechanisms and aimed to determine the effects of TCDD on upstream regulators of Pax5. A critical regulator of B cell differentiation, B lymphocyte-induced maturation protein-1 (Blimp-1) acts as a transcriptional repressor of Pax5. In lipopolysaccharide (LPS)-activated murine B cell lymphoma, CH12.LX, Blimp-1 messenger RNA, and DNA-binding activity within the Pax5 promoter were suppressed by TCDD. Furthermore, LPS activation of CH12.LX cells upregulated DNA-binding activity of activator protein 1 (AP-1) at three responsive element-like motifs within the Blimp-1 promoter. TCDD treatment of LPS-activated CH12.LX cells suppressed AP-1 binding to these motifs between 24 and 72 h, in concordance with the suppression of Blimp-1 by TCDD. A more comprehensive analysis at 72 h demonstrated that the suppression of AP-1 binding within the Blimp-1 promoter by TCDD was concentration dependent. In summary, our findings link the TCDD-mediated suppression of Blimp-1 through AP-1 to the dysregulation of Pax5, which ultimately leads to the suppression of B cell differentiation and humoral immune responses.


Toxicology and Applied Pharmacology | 2011

Regulation of Bach2 by the aryl hydrocarbon receptor as a mechanism for suppression of B-cell differentiation by 2,3,7,8-tetrachlorodibenzo-p-dioxin

K. Nadira De Abrew; Ashwini S. Phadnis; Robert B. Crawford; Norbert E. Kaminski; Russell S. Thomas

Exposure to the aryl hydrocarbon receptor (AHR) agonist, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) alters B-cell differentiation and suppresses antibody production. Previous genomic studies in mouse B cells identified Bach2 as a direct target of the AHR. Bach2 is known to repress expression of Prdm1, a key transcription factor involved in B-cell differentiation, by binding to Maf elements (MAREs) in the regulatory regions of the gene. Chromatin immunoprecipitation followed by quantitative PCR in TCDD-treated lipopolysaccharide (LPS)-activated B cells showed increased binding of the AHR within the first intron in the Bach2 gene. The binding was further confirmed by electrophoretic mobility shift assay (EMSA). TCDD also induced expression of Bach2 in activated as well as resting B cells from 2 to 24h post-treatment in a time- and concentration-dependent manner. Expression of Prdm1 was decreased by TCDD at 24h and was consistent with repression by Bach2. Increased DNA binding activity to the intron 5 MARE with increasing TCDD concentrations was observed by EMSA. Supershifts identified the presence of Bach2 in the DNA binding complex associated with the intron 5 MARE of Prdm1. Functional validation of the role of Bach2 in the suppression of B-cell differentiation by TCDD was performed using RNA interference (RNAi). Knockdown of Bach2 showed approximately 40% reversal in the TCDD-induced suppression of IgM secretion when compared to controls. The results suggest that the transcriptional regulation of Bach2 by the AHR is one of the mechanisms involved in the suppression of B-cell differentiation by TCDD.


BMC Systems Biology | 2010

Stochastic Modeling of B Lymphocyte Terminal Differentiation and Its Suppression by Dioxin

Qiang Zhang; Sudin Bhattacharya; Douglas E. Kline; Robert B. Crawford; Rory B. Conolly; Russell S. Thomas; Norbert E. Kaminski; Melvin E. Andersen

BackgroundUpon antigen encounter, naïve B lymphocytes differentiate into antibody-secreting plasma cells. This humoral immune response is suppressed by the environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and other dioxin-like compounds, which belong to the family of aryl hydrocarbon receptor (AhR) agonists.ResultsTo achieve a better understanding of the immunotoxicity of AhR agonists and their associated health risks, we have used computer simulations to study the behavior of the gene regulatory network underlying B cell terminal differentiation. The core of this network consists of two coupled double-negative feedback loops involving transcriptional repressors Bcl-6, Blimp-1, and Pax5. Bifurcation analysis indicates that the feedback network can constitute a bistable system with two mutually exclusive transcriptional profiles corresponding to naïve B cells and plasma cells. Although individual B cells switch to the plasma cell state in an all-or-none fashion when stimulated by the polyclonal activator lipopolysaccharide (LPS), stochastic fluctuations in gene expression make the switching event probabilistic, leading to heterogeneous differentiation response among individual B cells. Moreover, stochastic gene expression renders the dose-response behavior of a population of B cells substantially graded, a result that is consistent with experimental observations. The steepness of the dose response curve for the number of plasma cells formed vs. LPS dose, as evaluated by the apparent Hill coefficient, is found to be inversely correlated to the noise level in Blimp-1 gene expression. Simulations illustrate how, through AhR-mediated repression of the AP-1 protein, TCDD reduces the probability of LPS-stimulated B cell differentiation. Interestingly, stochastic simulations predict that TCDD may destabilize the plasma cell state, possibly leading to a reversal to the B cell phenotype.ConclusionOur results suggest that stochasticity in gene expression, which renders a graded response at the cell population level, may have been exploited by the immune system to launch humoral immune response of a magnitude appropriately tuned to the antigen dose. In addition to suppressing the initiation of the humoral immune response, dioxin-like compounds may also disrupt the maintenance of the acquired immunity.

Collaboration


Dive into the Robert B. Crawford's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jinpeng Li

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Weimin Chen

Michigan State University

View shared research outputs
Top Co-Authors

Avatar

Haitian Lu

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian J. Teppen

Michigan State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge