scholarly journals Dynamic Imaging of IEL-IEC Co-Cultures Allows for Quantification of CD103-Dependent T Cell Migration

2021 ◽  
Vol 22 (10) ◽  
pp. 5148
Author(s):  
Karin Enderle ◽  
Martin Dinkel ◽  
Eva-Maria Spath ◽  
Benjamin Schmid ◽  
Sebastian Zundler ◽  
...  

Intraepithelial lymphocytes (IEL) are widely distributed within the small intestinal epithelial cell (IEC) layer and represent one of the largest T cell pools of the body. While implicated in the pathogenesis of intestinal inflammation, detailed insight especially into the cellular cross-talk between IELs and IECs is largely missing in part due to lacking methodologies to monitor this interaction. To overcome this shortcoming, we employed and validated a murine IEL-IEC (organoids) ex vivo co-culture model system. Using livecell imaging we established a protocol to visualize and quantify the spatio-temporal migratory behavior of IELs within organoids over time. Applying this methodology, we found that IELs lacking CD103 (i.e., integrin alpha E, ITGAE) surface expression usually functioning as a retention receptor for IELs through binding to E-cadherin (CD324) expressing IECs displayed aberrant mobility and migration patterns. Specifically, CD103 deficiency affected the ability of IELs to migrate and reduced their speed during crawling within organoids. In summary, we report a new technology to monitor and quantitatively assess especially migratory characteristics of IELs communicating with IEC ex vivo. This approach is hence readily applicable to study the effects of targeted therapeutic interventions on IEL-IEC cross-talk.

Author(s):  
Bindu Chandrasekharan ◽  
Darra Boyer ◽  
Joshua A Owens ◽  
Alexandra A Wolfarth ◽  
Bejan J Saeedi ◽  
...  

Abstract We have demonstrated that neuropeptide Y (NPY) can regulate pro-inflammatory signaling in the gut via cross-talk with the pro-inflammatory cytokine tumor necrosis factor (TNF). Here, we investigated if selective blocking of NPY receptors, NPY1R or NPY2R, using small molecule non-peptide antagonists (BIBP-3222 for NPY1R and BIIE-0246 for NPY2R) in the colon could attenuate intestinal inflammation by lowering TNF levels (BIBP - N-[(1R)]-4-[(Aminoiminomethyl)amino-1-[[[(4-hydroxyphenyl)methyl]amino]carbonyl]butyl-α-phenylbenzeneacetamide; BIIE - N-[(1S)-4-[(Aminoiminomethyl)amino]-1-[[[2-(3,5-dioxo-1,2-diphenyl-1,2,4-triazolidin-4-yl)ethyl]amino]carbonyl]butyl]-1-[2-[4-(6,11-dihydro-6-oxo-5H-dibenz[b,e]azepin-11-yl)-1-piperazinyl]-2-oxoethyl]-cyclopentaneacetamide). Colitis was induced using dextran sodium sulfate in drinking water for 7 days, or by adoptive T-cell transfer in RAG-/- mice. Colonic biopsies from healthy subjects (n = 10) and IBD patients (n = 34, UC = 20, CD = 14) were cultured ex vivo in presence or absence of NPY antagonists (100 µM, 20 h), and cytokine release into culture supernatants was measured by ELISA. Intracolonic administration of BIBP (but not BIIE) significantly reduced clinical, endoscopic, and histological scores, and serum TNF, interleukin (IL)-6, and IL-12p70 in DSS colitis; it also significantly attenuated histological damage and serum IL-6 in T-cell colitis (P < .05). Intracolonic administration of BIBP significantly reduced TNF and interferon (IFN)-γ release from UC biopsies, whereas BIIE downregulated only IFN-γ (P < .05). BIBP significantly reduced TNF and interferon (IFN)-γ release from UC biopsies, whereas BIIE downregulated only IFN-γ (P < .05). Our data suggest a promising therapeutic value for NPY1R inhibition in alleviating intestinal inflammation in UC, possibly as enemas to IBD patients.


2002 ◽  
Vol 196 (10) ◽  
pp. 1355-1361 ◽  
Author(s):  
Sandra M. Hayes ◽  
Karen Laky ◽  
Dalal El-Khoury ◽  
Dietmar J. Kappes ◽  
B.J. Fowlkes ◽  
...  

The T cell antigen receptor complexes expressed on αβ and γδ T cells differ not only in their respective clonotypic heterodimers but also in the subunit composition of their CD3 complexes. The γδ T cell receptors (TCRs) expressed on ex vivo γδ T cells lack CD3δ, whereas αβ TCRs contain CD3δ. While this result correlates with the phenotype of CD3δ−/− mice, in which γδ T cell development is unaffected, it is inconsistent with the results of previous studies reporting that CD3δ is a component of the γδ TCR. Since earlier studies examined the subunit composition of γδ TCRs expressed on activated and expanded peripheral γδ T cells or γδ TCR+ intestinal intraepithelial lymphocytes, we hypothesized that activation and expansion may lead to changes in the CD3 subunit composition of the γδ TCR. Here, we report that activation and expansion do in fact result in the inclusion of a protein, comparable in mass and mobility to CD3δ, in the γδ TCR. Further analyses revealed that this protein is not CD3δ, but instead is a differentially glycosylated form of CD3γ. These results provide further evidence for a major difference in the subunit composition of αβ- and γδ TCR complexes and raise the possibility that modification of CD3γ may have important functional consequences in activated γδ T cells.


2019 ◽  
Vol 37 (8_suppl) ◽  
pp. 30-30
Author(s):  
Patrick C. Gedeon ◽  
Carter M. Suryadevara ◽  
Bryan D. Choi ◽  
John H. Sampson

30 Background: Activated T cells are known to traffic throughout the body including past the blood-brain barrier where they perform routine immune surveillance. Whether activated T cells can be used to enhance the efficacy and delivery of intravenously-administered, immunotherapeutic antibodies has yet to be explored. Methods: To examine efficacy, T cell migration and antibody delivery in vivo, the invasive murine glioma, CT-2A-EGFRvIII, was implanted orthotopically in human CD3 transgenic mice. Cohorts of mice were given vehicle or 1x107 non-specifically activated, syngeneic T cells intravenously. Beginning the subsequent day, groups were treated with daily intravenous infusions of human-CD3-binding, tumor-lysis-inducing bispecific antibody (hEGFRvIII-CD3 bi-scFv) or control bispecific antibody. To block T cell extravasation, cohorts received natalizumab or isotype control via intraperitoneal injection every other day beginning on the day of adoptive cell transfer. T cell migration was assessed using whole body bioluminescence imaging of activated T cells transduced to express firefly luciferase. Bispecific antibody biodistribution was assessed using PET-CT imaging of iodine-124 labeled antibody. Results: Following intravenous administration, ex vivo activated T cells tracked to invasive, syngeneic, orthotopic glioma, reaching maximal levels on average four days following adoptive transfer. Administration of ex vivo activated T cells enhanced bispecific antibody efficacy causing a statistically significant increase in survival (p = 0.007) with 80% long-term survivors. Treatment with the T cell extravasation blocking molecule natalizumab abrogated the increase in efficacy to levels observed in cohorts that did not receive adoptive transfer of activated T cells (p = 0.922). Pre-administration with ex vivo activated T cells produced a statistically significant increase in tumor penetrance of radiolabeled bispecific antibody (p = 0.023). Conclusions: Adoptive transfer of non-specifically activated T cells enhances the efficacy and tumor penetrance of intravenously-administered CD3-binding bispecific antibody.


Metabolites ◽  
2021 ◽  
Vol 11 (10) ◽  
pp. 672
Author(s):  
Michelle Saoi ◽  
Philip Britz-McKibbin

Metabolomics offers a hypothesis-generating approach for biomarker discovery in clinical medicine while also providing better understanding of the underlying mechanisms of chronic diseases. Clinical metabolomic studies largely rely on human biofluids (e.g., plasma, urine) as a more convenient specimen type for investigation. However, biofluids are non-organ specific reflecting complex biochemical processes throughout the body, which may complicate biochemical interpretations. For these reasons, tissue metabolomic studies enable deeper insights into aberrant metabolism occurring at the direct site of disease pathogenesis. This review highlights new advances in metabolomics for ex vivo analysis, as well as in situ imaging of tissue specimens, including diverse tissue types from animal models and human participants. Moreover, we discuss key pre-analytical and post-analytical challenges in tissue metabolomics for robust biomarker discovery with a focus on new methodological advances introduced over the past six years, including innovative clinical applications for improved screening, diagnostic testing, and therapeutic interventions for cancer.


2020 ◽  
Author(s):  
Khader Ghneim ◽  
Ashish A Sharma ◽  
Susan P Ribeiro ◽  
Slim Fourati ◽  
Jeffery Ahlers ◽  
...  

Current therapeutic interventions to eradicate latent HIV ("reservoir") and restore immune function in ART-treated HIV infection have yet to show efficacy. To explore mechanisms of HIV persistence, we apply an integrated systems biology approach and identify a distinct group of individuals with poor CD4 T-cell reconstitution (Immunologic non-responders, "INRs") and high frequencies of cells with inducible HIV. Contrary to the prevailing notion that immune activation drives HIV persistence and immune dysfunction, peripheral blood leukocytes from these subjects have enhanced expression of a network of genes regulated by cellular senescence driving transcription factors (TFs) FOXO3, SMAD2 and IRF3. In these subjects, increased frequencies of regulatory T cells and expression of the TGF-β signaling cascade are complimented by the downregulation of cell cycle, metabolic and pro-inflammatory pathways. Lactobacillaceae family and metabolites (members of the butyrate family - i.e. α-ketobutyrate) were correlated with Treg frequencies in "Senescent-INRs" ex vivo, triggered the differentiation of TGF-β producing Tregs and promoted HIV latency establishment in vitro. These cascades, downstream of PD-1/TGF-β prevent memory T cell differentiation and are associated with an increase in frequencies of cells with inducible HIV ex vivo. Our findings identify cellular senescence responses that can be targeted by PD-1 or TGF-β specific interventions that have shown safety and efficacy in cancer, and may prove to be crucial for HIV eradication.


2018 ◽  
Author(s):  
Alina Huth ◽  
Xiaoling Liang ◽  
Stefan Krebs ◽  
Helmut Blum ◽  
Andreas Moosmann

AbstractCytomegalovirus (CMV) is a prevalent human pathogen. The virus cannot be eliminated from the body, but is kept in check by CMV-specific T cells. Patients with an insufficient T-cell response, such as transplant recipients, are at high risk of developing CMV disease. However, the CMV-specific T-cell repertoire is complex, and is not yet clear which T cells protect best against virus reactivation and disease. Here we present a highly resolved characterization of CMV-specific CD8+ T cells based on enrichment by specific peptide stimulation and mRNA sequencing of their T-cell receptor β chains (TCRβ). Our analysis included recently identified T-cell epitopes restricted through HLA-C, whose presentation is resistant to viral immunomodulation, and well-studied HLA-B-restricted epitopes. In 8 healthy virus carriers, we identified a total of 1052 CMV-specific TCRβ chains. HLA-C-restricted, CMV-specific TCRβ clonotypes theex vivoT-cell response, and contributed the highest-frequency clonotype of the entire repertoire in 2 of 8 donors. We analyzed sharing and similarity of CMV-specific TCRβ sequences and identified 63 public or related sequences belonging to 17 public TCRβ families. In our cohort and in an independent cohort of 352 donors, the cumulative frequency of these public TCRβ family members was a highly discriminatory indicator of carrying both CMV infection and the relevant HLA type. Based on these findings, we propose CMV-specific TCRβ signatures as a biomarker for an antiviral T-cell response to identify patients in need of treatment and to guide future development of immunotherapy.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 43-43
Author(s):  
Patricia Taylor ◽  
Angela Panoskaltsis-Mortari ◽  
Randolph Noelle ◽  
Alexander Rudensky ◽  
Jonathan Serody ◽  
...  

Abstract The process by which bone marrow is rejected by host T cells has not been able to be directly visualized to date. To study the process of allogeneic bone marrow rejection and the effects of therapeutic interventions, we created 2 models that allow us to 1) quantify the specific expansion of host-type alloreactive T cells early post-transplant in response to allogeneic BM infusion and to 2) image host T cells in vivo during BM rejection. For the first model, 2C and TEa lymph node (LN) cells were adoptively transferred into syngeneic C57BL/6 (B6) Rag deficient mice on d-2. 2C CD8+ and TEa CD4+ T cell receptor transgenic T cells are reactive against BALB/c alloantigen. Mice were irradiated with 200 cGy on d-1 and BALB/c BM was infused on d0. Controls included mice that received 2C/TEa LN cells but no BM. Ten days later, spleen analysis revealed that 2C CD8+ and TEa CD4+ T cells had expanded 322-fold and 33-fold (ave of 6 exp.), respectively, in mice receiving BALB/c BM compared to controls that did not receive BM. Expanded T cells were activated as determined by flow cytometric parameters and cell surface antigens. Data indicate that host alloreactive T cell expansion was inhibited by >95% by combined, but not single, costimulatory pathway blockade. Studies are in progress to analyze in vitro host anti-donor responses of adoptively transferred T cells. To visualize the response of host T cells to donor BM in vivo, we developed a rejection model for imaging involving the adoptive transfer of green fluorescent protein (GFP) T cells (obtained from GFP transgenic mice) into syngeneic non-GFP B6 recipients immediately following sublethal irradiation (500 cGy). Allogeneic BALB/c or syngeneic B6 BM was infused the following day. The syngeneic BMT controls allowed for the distinction of homeostatic vs alloreactive expansion of GFP+ T cells. Transplanted mice not receiving GFP T cells served as negative controls to verify lack of autofluorescence. Cohorts were imaged d4 to d18 post BMT. By d4, low numbers of GFP+ cells were evident in femoral BM cavity, peripheral and mesenteric LNs, spleen, Peyer’s patches (PP) and to a lesser extent, lung. By d7, massive expansion of GFP+ cells could be visualized throughout the body of recipients of allogeneic BM. LNs, spleen, PP (peri-follicular area) and BM cavity increased dramatically in GFP intensity from d4 to d7. On d7 to d14, there were large foci of GFP+ cells in the lung, liver, skin, gingiva, kidney, uterus, and colon in allogeneic BMT recipients. Compared with allogeneic BMT recipients, syngeneic BMT recipients had greatly reduced numbers of GFP+ T cells in lymphoid organs and only rare cells were noted in liver, kidney, skin, BM and gingiva. In both allogeneic and syngeneic BMT recipients, lengths of ileum were diffusely infiltrated while other sections contained discrete foci of GFP+ cells. These imaging data provide a vivid illustration of the massive expansion and multi-organ distribution of host anti-donor T cells in vivo. Recent generation of GFP+ 2C and GFP+ TEa mice will permit the imaging of alloantigen-specific T cells during a rejection response. Additional imaging experiments are planned to study the fate of GFP+ BM transferred to allogeneic recipients under conditions of engraftment vs rejection. These models provide a unique platform for the testing of therapeutic interventions.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3888-3888
Author(s):  
Hugo De La Pena ◽  
J. Alejandro Madrigal ◽  
M. Bencsik ◽  
Gareth W.V. Cave ◽  
A. Selman ◽  
...  

Abstract T cells are probably one of the most pivotal cell types in the human adaptive immune system. They have the capability to eradicate primary, metastatic, relapsed tumours and can ameliorate otherwise fatal viral infections. Not surprisingly therefore, activation and expansion of T cells has become one of the main focuses for immunotherapy and immune gene therapy. Sufficient T cells numbers however, are required to deliver a significant clinical impact to patients, and rapid reproducible expansion of viable T cells still remains one of the main challenges for significant improvement. One of the main concerns with adoptive immunotherapy is that it relies on one critical factor: ex-vivo cell manipulation; the problem with this is that the longer the in-vitro T cell culture, the shorter the in-vivo T cell survival after infusion. In-vivo artificial expansion systems for active immunotherapy would clearly circumvent this problem. Therefore ideally a flexible system should be constructed in order to performed both adoptive and/or active immunotherapy depending on the patients requirements. Currently there is no comprehensive artificial Antigen Presenting Cell system (aAPC) for both effective ex-vivo and in-vivo antigen specific T cell expansion. In order to address this, using nanotechnology, we have constructed a nano sized super-para-magnetic artificial targeted and traceable in-vivo APC system by coating liposomes (approved for human use) with an optimised number of MHC Class I / peptide complexes and a specific selected range of ligands for adhesion (anti LFA1), early activation (anti CD28, anti CD27), late activation (anti 4-1BB) and survival (anti CD40L) T cell receptors in the form of Fab antibody regions. We have constructed targeted liposomes (immuno-liposomes), which are also traceable in-vivo via fluorescent and Magnetic Resonance Imaging (MRI). Ex-vivo (human) and in-vivo (animal) models have been investigated showing firstly that these super-para-magnetic immuno-liposomes circulate the body safely and facilitate their own focusing to specific organs, tumour sites or body areas by applying external magnetic attraction. Secondly, in a viral (CMV) antigen specific model and measured by relevant and irrelevant tetramers, the system is capable of activating and expanding antigen specific T cells at greater levels (200 fold) than standard methods from CMV positive (memory) individuals. The system has also been able to accomplish a small successful level of T cell priming from naive CMV negative individuals. The T cells are phenotypicaly relevant and fully functional in terms of degranulation and cytokine production when specifically challenged. As mechanisms of action, we have demonstrated that the system functions directly on T cells as micro APCs and also semi-directly on the surface of natural APCs following a similar exosomes kinetics. The system is generated in less that 48 hr. Once the aAPC is created and it remains viable and stable for 7 days minimum. We have established optimal conditions for an efficient artificial APC, which embodies a superior and controllable approach and platform with enormous potential for cancer nanotechnology and T cell mediated immunotherapy.


2020 ◽  
Vol 117 (29) ◽  
pp. 17166-17176 ◽  
Author(s):  
Wendy A. Goodman ◽  
Sarah M. Bedoyan ◽  
Hannah L. Havran ◽  
Brian Richardson ◽  
Mark J. Cameron ◽  
...  

Signaling of 17β-estradiol (estrogen) through its two nuclear receptors, α and β (ERα, ERβ), is an important mechanism of transcriptional regulation. Although ERs are broadly expressed by cells of the immune system, the mechanisms by which they modulate immune responses remain poorly understood. ERβ-specific signaling is reduced in patients with chronic inflammatory diseases, including systemic lupus erythematosus and inflammatory bowel disease, and our previous work suggests that dysregulation of ERβ-specific signaling contributes to enhanced intestinal inflammation in female SAMP/YitFC mice, a spontaneous model of Crohn’s disease-like ileitis. The present study builds on these prior observations to identify a nonredundant, immunoprotective role for ERβ-specific signaling in TGF-β–dependent regulatory T cell (Treg) differentiation. Using a strain of congenic SAMP mice engineered to lack global expression of ERβ, we observed dramatic, female-specific exacerbation of intestinal inflammation accompanied by significant reductions in intestinal Treg frequency and function. Impaired Treg suppression in the absence of ERβ was associated with aberrant overexpression ofTsc22d3(GILZ), a glucocorticoid-responsive transcription factor not normally expressed in mature Tregs, and ex vivo data reveal that forced overexpression of GILZ in mature Tregs inhibits their suppressive function. Collectively, our findings identify a pathway of estrogen-mediated immune regulation in the intestine, whereby homeostatic expression of ERβ normally functions to limit Treg-specific expression of GILZ, thereby maintaining effective immune suppression. Our data suggest that transcriptional cross-talk between glucocorticoid and steroid sex hormone signaling represents an important and understudied regulatory node in chronic inflammatory disease.


Sign in / Sign up

Export Citation Format

Share Document