Network


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

Hotspot


Dive into the research topics where Adebowale O. Bamidele is active.

Publication


Featured researches published by Adebowale O. Bamidele.


Journal of Immunology | 2011

Stromal Cell-Derived Factor-1 Signaling via the CXCR4-TCR Heterodimer Requires Phospholipase C-β3 and Phospholipase C-γ1 for Distinct Cellular Responses

Kimberly N. Kremer; Ian C. Clift; Alexander G. Miamen; Adebowale O. Bamidele; Nan Xin Qian; Troy D. Humphreys; Karen E. Hedin

The CXCR4 chemokine receptor is a G protein-coupled receptor that signals in T lymphocytes by forming a heterodimer with the TCR. CXCR4 and TCR functions are consequently highly cross regulated, affecting T cell immune activation, cytokine secretion, and T cell migration. The CXCR4-TCR heterodimer stimulates T cell migration and activation of the ERK MAPK and downstream AP-1–dependent cytokine transcription in response to stromal cell-derived factor-1 (SDF-1), the sole chemokine ligand of CXCR4. These responses require Gi-type G proteins as well as TCR ITAM domains and the ZAP70 tyrosine kinase, thus indicating that the CXCR4-TCR heterodimer signals to integrate G protein-coupled receptor-associated and TCR-associated signaling molecules in response to SDF-1. Yet, the phospholipase C (PLC) isozymes responsible for coupling the CXCR4-TCR heterodimer to distinct downstream cellular responses are incompletely characterized. In this study, we demonstrate that PLC activity is required for SDF-1 to induce ERK activation, migration, and CXCR4 endocytosis in human T cells. SDF-1 signaling via the CXCR4-TCR heterodimer uses PLC-β3 to activate the Ras-ERK pathway and increase intracellular calcium ion concentrations, whereas PLC-γ1 is dispensable for these outcomes. In contrast, PLC-γ1, but not PLC-β3, is required for SDF-1–mediated migration via a mechanism independent of LAT. These results increase understanding of the signaling mechanisms employed by the CXCR4-TCR heterodimer, characterize new roles for PLC-β3 and PLC-γ1 in T cells, and suggest that multiple PLCs may also be activated downstream of other chemokine receptors to distinctly regulate migration versus other signaling functions.


Molecular Pharmacology | 2014

β-Arrestin1 and Distinct CXCR4 Structures Are Required for Stromal Derived Factor-1 to Downregulate CXCR4 Cell-Surface Levels in Neuroblastoma

Ian C. Clift; Adebowale O. Bamidele; Christie Rodriguez-Ramirez; Kimberly N. Kremer; Karen E. Hedin

CXC chemokine receptor 4 (CXCR4) is a G protein–coupled receptor (GPCR) located on the cell surface that signals upon binding the chemokine stromal derived factor-1 (SDF-1; also called CXCL 12). CXCR4 promotes neuroblastoma proliferation and chemotaxis. CXCR4 expression negatively correlates with prognosis and drives neuroblastoma growth and metastasis in mouse models. All functions of CXCR4 require its expression on the cell surface, yet the molecular mechanisms that regulate CXCR4 cell-surface levels in neuroblastoma are poorly understood. We characterized CXCR4 cell-surface regulation in the related SH-SY5Y and SK-N-SH human neuroblastoma cell lines. SDF-1 treatment caused rapid down-modulation of CXCR4 in SH-SY5Y cells. Pharmacologic activation of protein kinase C similarly reduced CXCR4, but via a distinct mechanism. Analysis of CXCR4 mutants delineated two CXCR4 regions required for SDF-1 treatment to decrease cell-surface CXCR4 in neuroblastoma cells: the isoleucine-leucine motif at residues 328 and 329 and residues 343–352. In contrast, and unlike CXCR4 regulation in other cell types, serines 324, 325, 338, and 339 were not required. Arrestin proteins can bind and regulate GPCR cell-surface expression, often functioning together with kinases such as G protein–coupled receptor kinase 2 (GRK2). Using SK-N-SH cells which are naturally deficient in β-arrestin1, we showed that β-arrestin1 is required for the CXCR4 343–352 region to modulate CXCR4 cell-surface expression following treatment with SDF-1. Moreover, GRK2 overexpression enhanced CXCR4 internalization, via a mechanism requiring both β-arrestin1 expression and the 343–352 region. Together, these results characterize CXCR4 structural domains and β-arrestin1 as critical regulators of CXCR4 cell-surface expression in neuroblastoma. β-Arrestin1 levels may therefore influence the CXCR4-driven metastasis of neuroblastoma as well as prognosis.


Molecular Pharmacology | 2014

β-arrestin1 and distinct CXCR4 structures are required for SDF-1 to down-regulate CXCR4 cell-surface levels in neuroblastoma

Ian C. Clift; Adebowale O. Bamidele; Christie Rodriguez-Ramirez; Kimberly N. Kremer; Karen E. Hedin

CXC chemokine receptor 4 (CXCR4) is a G protein–coupled receptor (GPCR) located on the cell surface that signals upon binding the chemokine stromal derived factor-1 (SDF-1; also called CXCL 12). CXCR4 promotes neuroblastoma proliferation and chemotaxis. CXCR4 expression negatively correlates with prognosis and drives neuroblastoma growth and metastasis in mouse models. All functions of CXCR4 require its expression on the cell surface, yet the molecular mechanisms that regulate CXCR4 cell-surface levels in neuroblastoma are poorly understood. We characterized CXCR4 cell-surface regulation in the related SH-SY5Y and SK-N-SH human neuroblastoma cell lines. SDF-1 treatment caused rapid down-modulation of CXCR4 in SH-SY5Y cells. Pharmacologic activation of protein kinase C similarly reduced CXCR4, but via a distinct mechanism. Analysis of CXCR4 mutants delineated two CXCR4 regions required for SDF-1 treatment to decrease cell-surface CXCR4 in neuroblastoma cells: the isoleucine-leucine motif at residues 328 and 329 and residues 343–352. In contrast, and unlike CXCR4 regulation in other cell types, serines 324, 325, 338, and 339 were not required. Arrestin proteins can bind and regulate GPCR cell-surface expression, often functioning together with kinases such as G protein–coupled receptor kinase 2 (GRK2). Using SK-N-SH cells which are naturally deficient in β-arrestin1, we showed that β-arrestin1 is required for the CXCR4 343–352 region to modulate CXCR4 cell-surface expression following treatment with SDF-1. Moreover, GRK2 overexpression enhanced CXCR4 internalization, via a mechanism requiring both β-arrestin1 expression and the 343–352 region. Together, these results characterize CXCR4 structural domains and β-arrestin1 as critical regulators of CXCR4 cell-surface expression in neuroblastoma. β-Arrestin1 levels may therefore influence the CXCR4-driven metastasis of neuroblastoma as well as prognosis.


Journal of Biological Chemistry | 2017

The Role of the Histone Methyltransferase Enhancer of Zeste Homolog 2 (EZH2) in the Pathobiological Mechanisms Underlying Inflammatory Bowel Disease (IBD)

Olga F. Sarmento; Phyllis A. Svingen; Yuning Xiong; Zhifu Sun; Adebowale O. Bamidele; Angela Mathison; Thomas C. Smyrk; Asha Nair; Michelle Gonzalez; Mary R. Sagstetter; Saurabh Baheti; Dermot P. McGovern; Jessica Friton; Konstantinos A. Papadakis; Goel Gautam; Ramnik J. Xavier; Raul Urrutia; William A. Faubion

Regulatory T (Treg) cells expressing the transcription factor FOXP3 play a pivotal role in maintaining immunologic self-tolerance. We and others have shown previously that EZH2 is recruited to the FOXP3 promoter and its targets in Treg cells. To further address the role for EZH2 in Treg cellular function, we have now generated mice that lack EZH2 specifically in Treg cells (EZH2Δ/ΔFOXP3+). We find that EZH2 deficiency in FOXP3+ T cells results in lethal multiorgan autoimmunity. We further demonstrate that EZH2Δ/ΔFOXP3+ T cells lack a regulatory phenotype in vitro and secrete proinflammatory cytokines. Of special interest, EZH2Δ/ΔFOXP3+ mice develop spontaneous inflammatory bowel disease. Guided by these results, we assessed the FOXP3 and EZH2 gene networks by RNA sequencing in isolated intestinal CD4+ T cells from patients with Crohns disease. Gene network analysis demonstrates that these CD4+ T cells display a Th1/Th17-like phenotype with an enrichment of gene targets shared by FOXP3 and EZH2. Combined, these results suggest that the inflammatory milieu found in Crohns disease could lead to or result from deregulation of FOXP3/EZH2-enforced T cell gene networks contributing to the underlying intestinal inflammation.


PLOS ONE | 2014

Coactosin-Like 1 Antagonizes Cofilin to Promote Lamellipodial Protrusion at the Immune Synapse

Joanna Kim; Michael J. Shapiro; Adebowale O. Bamidele; Pinar S. Gurel; Puspa Thapa; Henry N. Higgs; Karen E. Hedin; Virginia Smith Shapiro; Daniel D. Billadeau

Actin depolymerizing factor-homology (ADF-H) family proteins regulate actin filament dynamics at multiple cellular locations. Herein, we have investigated the function of the ADF-H family member coactosin-like 1 (COTL1) in the regulation of actin dynamics at the T cell immune synapse (IS). We initially identified COTL1 in a genetic screen to identify novel regulators of T cell activation, and subsequently found that it associates with F-actin and localizes at the IS in response to TCR+CD28 stimulation. Live cell microscopy showed that depletion of COTL1 protein impaired T cell spreading in response to TCR ligation and abrogated lamellipodial protrusion at the T cell – B cell contact site, producing only a band of F-actin. Significantly, re-expression of wild type COTL1, but not a mutant deficient in F-actin binding could rescue these defects. In addition, COTL1 depletion reduced T cell migration. In vitro studies showed that COTL1 and cofilin compete with each other for binding to F-actin, and COTL1 protects F-actin from cofilin-mediated depolymerization. While depletion of cofilin enhanced F-actin assembly and lamellipodial protrusion at the IS, concurrent depletion of both COTL1 and cofilin restored lamellipodia formation. Taken together, our results suggest that COTL1 regulates lamellipodia dynamics in part by protecting F-actin from cofilin-mediated disassembly.


Journal of Cell Biology | 2015

IQGAP1 promotes CXCR4 chemokine receptor function and trafficking via EEA-1+ endosomes

Adebowale O. Bamidele; Kimberly N. Kremer; Petra Hirsova; Ian C. Clift; Gregory J. Gores; Daniel D. Billadeau; Karen E. Hedin

IQGAP1 mediates CXCR4 cell surface expression and signaling by regulating EEA-1+ endosome interactions with microtubules during CXCR4 trafficking and recycling.


Inflammatory Bowel Diseases | 2016

O-015 YI Alterations in the FOXP3-EZH2 Pathway Associates with Increased Susceptibility to Colitis in Both Mice and Human.

Olga F. Sarmento; Yuning Xiong; Zhifu Sun; Phyllis A. Svingen; Adebowale O. Bamidele; Thomas C. Smyrk; Asha Nair; Saurabh Baheti; Dermot P. McGovern; Jessica Friton; Konstantinos A. Papadakis; Gautam Goel; Ramnik J. Xavier; Raul Urrutia; William A. Faubion

Background:Crohns disease is a common intestinal inflammatory disorder of uncertain etiology and incomplete treatment options. It is characterized by lesions infiltrated by inflammatory CD4+ lymphocytes; yet the mechanism of CD4+ mediated pathophysiology is unclear. Through whole genome approaches on clinical cohorts of CD patients, combined with functional in-vivo and in-vitro murine data, we sought to identify and evaluate aberrant transcriptional gene networks in disease-associated CD4+ cells. Methods:We comparatively studied the expression profile in CD4+ lymphocytes isolated from the Ileum of 21 CD-affected individuals and 12 age/gender matched control individuals. Utilizing RNA-seq, we conducted upstream target analysis to identify top disease associated regulatory networks. Potential coordinated function between FOXP3 and EZH2 was identified, and common gene targets were interrogated for differential expression and pathway analysis.We crossed FOXP3-driven CRE recombinase expressing mice (B6129S-Tg(Foxp3-EGFP/cre)1aJbs/J) with EZH2fl/fl animals to better define the effect of EZH2 ablation on Treg function. Splenocytes were analyzed by cell surface markers for T cell distribution and phenotype. EZH2&Dgr;/&Dgr; FOXP3+ cell function was determined by in-vitro Treg suppressor function assay. Cytokine analyses were performed on supernatants of stimulated EZH2&Dgr;/&Dgr;FOXP3+ splenocytes and serum from 14 to 17 days old FOXP3-CRE;EZH2&Dgr;/&Dgr; mice. FOXP3-CRE;EZH2&Dgr;/&Dgr; or animals treated with EZH2 inhibitors received 3% DSS in an acute model of colitis. Furthermore, RB45high transfer utilizing WT or EZH2&Dgr;/&Dgr;Tregs was used in a chronic model of colitis. Results:Differential expression analysis between CD and CTRL samples revealed 5328 statistically significant Differentially Expressed Genes (DEGs). Principle component analysis, and hierarchical clustering, separated patients from control subjects. Among the DEGs, 83 were well-defined transcription factors (TFs) identified by Master Regulatory Analysis. The transcription factor FOXP3 and its associated regulatory network emerged as a highly differentially expressed pathway in diseased samples by Master Regulatory Analysis, Ingenuity Pathway Analysis, and Gene Set Enrichment Analysis. Additionally, common gene targets for both FOXP3 and EZH2 were concurrently up regulated in disease. Genetic ablation of EZH2 in mouse FOXP3+ cells resulted in Treg cells converted to a TH1/TH17- like effector phenotype, a pattern shared by disease associated CD4+ T cells. These cells exhibited pro-inflammatory cell surface markers, cytokine expression, and reduced in-vitro suppressive capacity. EZH2&Dgr;/&Dgr; Tregs failed to mitigate both DSS and T cell mediated colitis, and mice treated with the EZH2 inhibitor DZNep were also more susceptible to DSS colitis. Conclusions:In conclusion, we compare EZH2/Foxp3 co-regulation of gene networks between Crohns lesions and controls, and in multiple mouse models, to clarify the role of epigenetic regulation in this disease process. Multiple colitis disease models show us that interfering with EZH2 by genetic or pharmaceutical methods results in increased susceptibility to colitis. We postulate the inflammatory milieu found in IBD modulates EZH2 function, thereby making it less able to co-repress non-Treg differentiation programs even in the presence of upregulated FOXP3. Further investigation into EZH2 mutations, signaling cascades and relevant post-translation modifications to the FOXP3-EZH2 gene network may assist to elucidate and predict disease progression and/or severity.


Epigenetics | 2018

Epigenetics of gastrointestinal diseases: notes from a workshop

David L. Marks; Rachel L. Olson; Raul Urrutia; Daniel D. Billadeau; Nilotpal Roy; George A. Calin; Muller Fabbri; Marina Koutsioumpa; Dimitrios Iliopoulos; Tamas Ordog; Robert C. Huebert; Olga F. Sarmento; Adebowale O. Bamidele; William A. Faubion; Gwen Lomberk; Jens T. Siveke; Nita Ahuja; Juan L. Iovanna; Ryan A. Hlady; Keith D. Robertson; John B. Kisiel; Christopher L. Pin; Martin E. Fernandez-Zapico

ABSTRACT International experts gathered at the Mayo Clinic (Rochester MN, USA) on February 27th-28th, 2017 for a meeting entitled ‘Basic and Translational Facets of the Epigenetics of GI Diseases’. This workshop summarized recent advances on the role of epigenetics in the pathobiology of gastrointestinal (GI) diseases. Highlights of the meeting included recent advances on the involvement of different epigenetic mechanisms in malignant and nonmalignant GI disorders and the epigenetic heterogeneity exhibited in these diseases. The translational value of epigenetic drugs, as well as the current and future use of epigenetic changes (i.e., DNA methylation patterns) as biomarkers for early detection tools or disease stratification were also important topics of discussion.


Cellular and molecular gastroenterology and hepatology | 2018

Disruption of FOXP3–EZH2 Interaction Represents a Pathobiological Mechanism in Intestinal Inflammation

Adebowale O. Bamidele; Phyllis A. Svingen; Mary R. Sagstetter; Olga F. Sarmento; Michelle Gonzalez; Manuel B. Braga Neto; Subra Kugathasan; Gwen Lomberk; Raul Urrutia; William A. Faubion

Background & Aims Forkhead box protein 3 (FOXP3)+ regulatory T cell (Treg) dysfunction is associated with autoimmune diseases; however, the mechanisms responsible for inflammatory bowel disease pathophysiology are poorly understood. Here, we tested the hypothesis that a physical interaction between transcription factor FOXP3 and the epigenetic enzyme enhancer of zeste homolog 2 (EZH2) is essential for gene co-repressive function. Methods Human FOXP3 mutations clinically relevant to intestinal inflammation were generated by site-directed mutagenesis. T lymphocytes were isolated from mice, human blood, and lamina propria of Crohn’s disease (CD) patients and non-CD controls. We performed proximity ligation or a co-immunoprecipitation assay in FOXP3-mutant+, interleukin 6 (IL6)-treated or CD-CD4+ T cells to assess FOXP3–EZH2 protein interaction. We studied IL2 promoter activity and chromatin state of the interferon γ locus via luciferase reporter and chromatin-immunoprecipitation assays, respectively, in cells expressing FOXP3 mutants. Results EZH2 binding was abrogated by inflammatory bowel disease–associated FOXP3 cysteine 232 (C232) mutation. The C232 mutant showed impaired repression of IL2 and diminished EZH2-mediated trimethylation of histone 3 at lysine 27 on interferon γ, indicative of compromised Treg physiologic function. Generalizing this mechanism, IL6 impaired FOXP3–EZH2 interaction. IL6-induced effects were reversed by Janus kinase 1/2 inhibition. In lamina propria–derived CD4+T cells from CD patients, we observed decreased FOXP3–EZH2 interaction. Conclusions FOXP3–C232 mutation disrupts EZH2 recruitment and gene co-repressive function. The proinflammatory cytokine IL6 abrogates FOXP3–EZH2 interaction. Studies in lesion-derived CD4+ T cells have shown that reduced FOXP3–EZH2 interaction is a molecular feature of CD patients. Destabilized FOXP3–EZH2 protein interaction via diverse mechanisms and consequent Treg abnormality may drive gastrointestinal inflammation.


Gastroenterology | 2018

15 DISRUPTION OF FOXP3-EZH2 INTERACTION BY GENETIC MUTATION OR SIGNALING-INDUCED INACTIVATION REPRESENTS A NOVEL PATHOBIOLOGICAL MECHANISM IN INFLAMMATORY BOWEL DISEASE

Adebowale O. Bamidele; Phyllis A. Svingen; Mary R. Sagstetter; Michelle Gonzalez; Olga F. Sarmento; Manuel B. Braga Neto; Jessica Friton; Subra Kugathasan; Gwen Lomberk; Raul Urrutia; William A. Faubion

Collaboration


Dive into the Adebowale O. Bamidele's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge