Network


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

Hotspot


Dive into the research topics where Samuel Bertin is active.

Publication


Featured researches published by Samuel Bertin.


Nature Immunology | 2014

The ion channel TRPV1 regulates the activation and proinflammatory properties of CD4+ T cells

Samuel Bertin; Yukari Aoki-Nonaka; Petrus R. de Jong; Lilian Nohara; Hongjian Xu; Shawna Stanwood; Sonal Srikanth; Jihyung Lee; Keith To; Lior Abramson; Timothy Yu; Tiffany Han; Ranim Touma; Xiangli Li; José M. González-Navajas; Scott Herdman; Maripat Corr; Guo Fu; Hui Dong; Yousang Gwack; Alessandra Franco; Wilfred A. Jefferies; Eyal Raz

TRPV1 is a Ca2+-permeable channel studied mostly as a pain receptor in sensory neurons. However, its role in other cell types is poorly understood. Here we found that TRPV1 was functionally expressed in CD4+ T cells, where it acted as a non–store-operated Ca2+ channel and contributed to T cell antigen receptor (TCR)-induced Ca2+ influx, TCR signaling and T cell activation. In models of T cell–mediated colitis, TRPV1 promoted colitogenic T cell responses and intestinal inflammation. Furthermore, genetic and pharmacological inhibition of TRPV1 in human CD4+ T cells recapitulated the phenotype of mouse Trpv1−/− CD4+ T cells. Our findings suggest that inhibition of TRPV1 could represent a new therapeutic strategy for restraining proinflammatory T cell responses.


Journal of Clinical Investigation | 2012

Divergent requirement for Gαs and cAMP in the differentiation and inflammatory profile of distinct mouse Th subsets.

Xiangli Li; Fiona Murray; Naoki Koide; Jonathan Goldstone; Sara M. Dann; Jianzhong Chen; Samuel Bertin; Guo Fu; Lee S. Weinstein; Min Chen; Maripat Corr; Lars Eckmann; Paul A. Insel; Eyal Raz

cAMP, the intracellular signaling molecule produced in response to GPCR signaling, has long been recognized as an immunosuppressive agent that inhibits T cell receptor activation and T cell function. However, recent studies show that cAMP also promotes T cell-mediated immunity. Central to cAMP production downstream of GPCR activation is the trimeric G protein Gs. In order to reconcile the reports of divergent effects of cAMP in T cells and to define the direct effect of cAMP in T cells, we engineered mice in which the stimulatory Gα subunit of Gs (Gαs) could be deleted in T cells using CD4-Cre (Gnas(ΔCD4)). Gnas(ΔCD4) CD4(+) T cells had reduced cAMP accumulation and Ca2(+) influx. In vitro and in vivo, Gnas(ΔCD4) CD4(+) T cells displayed impaired differentiation to specific Th subsets: Th17 and Th1 cells were reduced or absent, but Th2 and regulatory T cells were unaffected. Furthermore, Gnas(ΔCD4) CD4(+) T cells failed to provoke colitis in an adoptive transfer model, indicating reduced inflammatory function. Restoration of cAMP levels rescued the impaired phenotype of Gnas(ΔCD4) CD4(+) T cells, reinstated the PKA-dependent influx of Ca2(+), and enhanced the ability of these cells to induce colitis. Our findings thus define an important role for cAMP in the differentiation of Th subsets and their subsequent inflammatory responses, and provide evidence that altering cAMP levels in CD4(+) T cells could provide an immunomodulatory approach targeting specific Th subsets.


Mucosal Immunology | 2015

TRPM8 on mucosal sensory nerves regulates colitogenic responses by innate immune cells via CGRP

P. R. De Jong; Naoko Takahashi; Madusha Peiris; Samuel Bertin; Jongdae Lee; Mélanie G. Gareau; A. Paniagua; Alexandra R. Harris; D. S. Herdman; Maripat Corr; L.A. Blackshaw; Eyal Raz

TRPM8 is the molecular sensor for cold; however, the physiological role of TRPM8+ neurons at mucosal surfaces is unclear. Here we evaluated the distribution and peptidergic properties of TRPM8+ fibers in naive and inflamed colons, as well as their role in mucosal inflammation. We found that Trpm8−/− mice were hypersusceptible to dextran sodium sulfate (DSS)-induced colitis, and that Trpm8−/− CD11c+ DCs (dendritic cells) showed hyperinflammatory responses to toll-like receptor (TLR) stimulation. This was phenocopied in calcitonin gene–related peptide (CGRP) receptor-deficient mice, but not in substance P receptor-deficient mice, suggesting a functional link between TRPM8 and CGRP. The DSS phenotype of CGRP receptor-deficient mice could be adoptively transferred to wild-type (WT) mice, suggesting that CGRP suppresses the colitogenic activity of bone marrow–derived cells. TRPM8+ mucosal fibers expressed CGRP in human and mouse colon. Furthermore, neuronal CGRP contents were increased in colons from naive and DSS-treated Trpm8−/− mice, suggesting deficient CGRP release in the absence of TRPM8 triggering. Finally, treatment of Trpm8−/− mice with CGRP reversed their hyperinflammatory phenotype. These results suggest that TRPM8 signaling in mucosal sensory neurons is indispensable for the regulation of innate inflammatory responses via the neuropeptide CGRP.


Journal of Clinical Investigation | 2014

Ion channel TRPV1-dependent activation of PTP1B suppresses EGFR-associated intestinal tumorigenesis

Petrus R. de Jong; Naoki Takahashi; Alexandra R. Harris; Jihyung Lee; Samuel Bertin; James Jeffries; Michael Jung; Jen Duong; Amy Triano; Jongdae Lee; Yaron Niv; David S. Herdman; Koji Taniguchi; Chang-Whan Kim; Hui Dong; Lars Eckmann; Stephanie M. Stanford; Nunzio Bottini; Maripat Corr; Eyal Raz

The intestinal epithelium has a high rate of turnover, and dysregulation of pathways that regulate regeneration can lead to tumor development; however, the negative regulators of oncogenic events in the intestinal epithelium are not fully understood. Here we identified a feedback loop between the epidermal growth factor receptor (EGFR), a known mediator of proliferation, and the transient receptor potential cation channel, subfamily V, member 1 (TRPV1), in intestinal epithelial cells (IECs). We found that TRPV1 was expressed by IECs and was intrinsically activated upon EGFR stimulation. Subsequently, TRPV1 activation inhibited EGFR-induced epithelial cell proliferation via activation of Ca2+/calpain and resulting activation of protein tyrosine phosphatase 1B (PTP1B). In a murine model of multiple intestinal neoplasia (Apc(Min/+) mice), TRPV1 deficiency increased adenoma formation, and treatment of these animals with an EGFR kinase inhibitor reversed protumorigenic phenotypes, supporting a functional association between TRPV1 and EGFR signaling in IECs. Administration of a TRPV1 agonist suppressed intestinal tumorigenesis in Apc(Min/+) mice, similar to--as well as in conjunction with--a cyclooxygenase-2 (COX-2) inhibitor, which suggests that targeting both TRPV1 and COX-2 has potential as a therapeutic approach for tumor prevention. Our findings implicate TRPV1 as a regulator of growth factor signaling in the intestinal epithelium through activation of PTP1B and subsequent suppression of intestinal tumorigenesis.


Nature Communications | 2016

ERK5 signalling rescues intestinal epithelial turnover and tumour cell proliferation upon ERK1/2 abrogation.

Petrus R. de Jong; Koji Taniguchi; Alexandra R. Harris; Samuel Bertin; Naoki Takahashi; Jen Duong; Alejandro D. Campos; Garth Powis; Maripat Corr; Michael Karin; Eyal Raz

The ERK1/2 MAPK signalling module integrates extracellular cues that induce proliferation and differentiation of epithelial lineages, and is an established oncogenic driver, particularly in the intestine. However, the interrelation of the ERK1/2 module relative to other signalling pathways in intestinal epithelial cells and colorectal cancer (CRC) is unclear. Here we show that loss of Erk1/2 in intestinal epithelial cells results in defects in nutrient absorption, epithelial cell migration and secretory cell differentiation. However, intestinal epithelial cell proliferation is not impeded, implying compensatory mechanisms. Genetic deletion of Erk1/2 or pharmacological targeting of MEK1/2 results in supraphysiological activity of the ERK5 pathway. Furthermore, targeting both pathways causes a more effective suppression of cell proliferation in murine intestinal organoids and human CRC lines. These results suggest that ERK5 provides a common bypass route in intestinal epithelial cells, which rescues cell proliferation upon abrogation of ERK1/2 signalling, with therapeutic implications in CRC.


Mucosal Immunology | 2015

Dual-specificity phosphatase 6 regulates CD4+ T-cell functions and restrains spontaneous colitis in IL-10-deficient mice.

Samuel Bertin; Beatriz Lozano-Ruiz; Victoria Bachiller; Irma García-Martínez; Scott Herdman; Pedro Zapater; Rubén Francés; José Such; Jongdae Lee; Eyal Raz; José M. González-Navajas

Mitogen-activated protein kinase (MAPK) phosphatases are dual-specificity phosphatases (DUSPs) that dephosphorylate phosphothreonine and phosphotyrosine residues within MAPKs. DUSP6 preferentially dephosphorylates extracellular signal-regulated kinases 1 and 2 (ERK1/2) rendering them inactive. Here, we study the role of DUSP6 in CD4+ T-cell function, differentiation, and inflammatory profile in the colon. Upon T-cell receptor (TCR) stimulation, DUSP6 knockout (Dusp6−/−) CD4+ T cells showed increased ERK1/2 activation, proliferation, T helper 1 differentiation, and interferon-γ production, as well as a marked decrease in survival, interleukin- 17A (IL-17A) secretion, and regulatory T-cell function. To analyze the role of DUSP6 in vivo, we employed the Il10−/− model of colitis and generated Il10−/−/Dusp6−/− double-knockout mice. Il10−/−/Dusp6−/− mice suffered from accelerated and exacerbated spontaneous colitis, which was prevented by ERK1/2 inhibition. ERK1/2 inhibition also augmented regulatory T-cell differentiation in vitro and in vivo in both C57Bl/6 and Dusp6−/− mice. In summary, DUSP6 regulates CD4+ T-cell activation and differentiation by inhibiting the TCR-dependent ERK1/2 activation. DUSP6 might therefore be a potential intervention target for limiting aberrant T-cell responses in T-cell-mediated diseases, such as inflammatory bowel disease.


Gut | 2017

The TRPA1 ion channel is expressed in CD4+ T cells and restrains T-cell-mediated colitis through inhibition of TRPV1

Samuel Bertin; Yukari Aoki-Nonaka; Jihyung Lee; Petrus R. de Jong; Peter Kim; Tiffany Han; Timothy Yu; Keith To; Naoki Takahashi; Brigid S. Boland; John T. Chang; Samuel B. Ho; Scott Herdman; Maripat Corr; Alessandra Franco; Sonia Sharma; Hui Dong; Armen N. Akopian; Eyal Raz

Objective Transient receptor potential ankyrin-1 (TRPA1) and transient receptor potential vanilloid-1 (TRPV1) are calcium (Ca2+)-permeable ion channels mostly known as pain receptors in sensory neurons. However, growing evidence suggests their crucial involvement in the pathogenesis of IBD. We explored the possible contribution of TRPA1 and TRPV1 to T-cell-mediated colitis. Design We evaluated the role of Trpa1 gene deletion in two models of experimental colitis (ie, interleukin-10 knockout and T-cell-adoptive transfer models). We performed electrophysiological and Ca2+ imaging studies to analyse TRPA1 and TRPV1 functions in CD4+ T cells. We used genetic and pharmacological approaches to evaluate TRPV1 contribution to the phenotype of Trpa1−/− CD4+ T cells. We also analysed TRPA1 and TRPV1 gene expression and TRPA1+TRPV1+ T cell infiltration in colonic biopsies from patients with IBD. Results We identified a protective role for TRPA1 in T-cell-mediated colitis. We demonstrated the functional expression of TRPA1 on the plasma membrane of CD4+ T cells and identified that Trpa1−/− CD4+ T cells have increased T-cell receptor-induced Ca2+ influx, activation profile and differentiation into Th1-effector cells. This phenotype was abrogated upon genetic deletion or pharmacological inhibition of the TRPV1 channel in mouse and human CD4+ T cells. Finally, we found differential regulation of TRPA1 and TRPV1 gene expression as well as increased infiltration of TRPA1+TRPV1+ T cells in the colon of patients with IBD. Conclusions Our study indicates that TRPA1 inhibits TRPV1 channel activity in CD4+ T cells, and consequently restrains CD4+ T-cell activation and colitogenic responses. These findings may therefore have therapeutic implications for human IBD.


Seminars in Immunopathology | 2016

Transient Receptor Potential (TRP) channels in T cells

Samuel Bertin; Eyal Raz

The transient receptor potential (TRP) family of ion channels is widely expressed in many cell types and plays various physiological roles. Growing evidence suggests that certain TRP channels are functionally expressed in the immune system. Indeed, an increasing number of reports have demonstrated the functional expression of several TRP channels in innate and adaptive immune cells and have highlighted their critical role in the activation and function of these cells. However, very few reviews have been entirely dedicated to this subject. Here, we will summarize the recent findings with regards to TRP channel expression in T cells and discuss their emerging role as regulators of T cell activation and functions. Moreover, these studies suggest that beyond their pharmaceutical interest in pain management, certain TRP channels may represent potential novel therapeutic targets for various immune-related diseases.


Scientific Reports | 2016

Neuronal TRPV1 activation regulates alveolar bone resorption by suppressing osteoclastogenesis via CGRP

Naoki Takahashi; Yumi Matsuda; Keisuke Sato; Petrus R. de Jong; Samuel Bertin; Koichi Tabeta; Kazuhisa Yamazaki

The transient receptor potential vanilloid 1 (TRPV1) channel is abundantly expressed in peripheral sensory neurons where it acts as an important polymodal cellular sensor for heat, acidic pH, capsaicin, and other noxious stimuli. The oral cavity is densely innervated by afferent sensory neurons and is a highly specialized organ that protects against infections as well as physical, chemical, and thermal stresses in its capacity as the first part of the digestive system. While the function of TRPV1 in sensory neurons has been intensively studied in other organs, its physiological role in periodontal tissues is unclear. In this study we found that Trpv1−/− mice developed severe bone loss in an experimental model of periodontitis. Chemical ablation of TRPV1-expressing sensory neurons recapitulated the phenotype of Trpv1−/− mice, suggesting a functional link between neuronal TRPV1 signaling and periodontal bone loss. TRPV1 activation in gingival nerves induced production of the neuropeptide, calcitonin gene-related peptide (CGRP), and CGRP treatment inhibited osteoclastogenesis in vitro. Oral administration of the TRPV1 agonist, capsaicin, suppressed ligature-induced bone loss in mice with fewer tartrate-resistant acid phosphatase (TRAP)-positive cells in alveolar bone. These results suggest that neuronal TRPV1 signaling in periodontal tissue is crucial for the regulation of osteoclastogenesis via the neuropeptide CGRP.


Channels | 2014

Novel immune function for the TRPV1 channel in T lymphocytes

Samuel Bertin; Petrus R. de Jong; Wilfred A. Jefferies; Eyal Raz

Novel immune function for the TRPV1 channel in T lymphocytes Samuel Bertin, Petrus Rudolf de Jong, Wilfred A Jefferies & Eyal Raz a Department of Medicine, UCSD; La Jolla, CA USA b Sanford-Burnham Medical Research Institute (SBMRI); La Jolla, CA USA c Michael Smith Laboratories, Center for Blood Research; The Brain Research Center; Department of Medical Genetics: Department of Microbiology and Immunology and Department of Zoology; University of British Columbia; Vancouver, Canada Accepted author version posted online: 21 Dec 2014.Published online: 23 Jan 2015.

Collaboration


Dive into the Samuel Bertin's collaboration.

Top Co-Authors

Avatar

Eyal Raz

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maripat Corr

University of California

View shared research outputs
Top Co-Authors

Avatar

Scott Herdman

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hui Dong

University of California

View shared research outputs
Top Co-Authors

Avatar

Jihyung Lee

University of California

View shared research outputs
Top Co-Authors

Avatar

Jongdae Lee

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guo Fu

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge