An Ear-Tissue Model for High-Resolution In Vivo Imaging.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1456-1456
Author(s):  
Yiqun Jiao ◽  
Ping Zhang ◽  
Divino DeOliveira ◽  
Nicholas Drago ◽  
Nelson J. Chao ◽  
...  

Abstract Abstract 1456 The use of microscopy in medicine has revolutionized medical research, diagnosis, and treatment. Unfortunately, current use of microscopy is mostly limited to 2 dimensional structures. The introduction of next generation microscopy, such as confocal and multiphoton, has enabled study of 3 dimensional structures deep in living tissues. Because there is a limit as to how deep these microscopic techniques can detect signal in tissue, many tissues/organs can not be studied due to inaccessibility (e.g., thymus). We have been using an ear-heart murine model to study immune tolerance. In this model, a heart from a newborn mouse is transplanted subcutaneously into the ear pinna. If the heart is not rejected (e.g., syngeneic setting), it can survive and beat indefinitely. In this study, we tested a hypothesis that other tissues can also be transplanted into the mouse ear pinna and function. Skin on the mouse ear pinna is extremely thin (<15 μm), thus allowing for visualization of cellular and subcellular changes in transplanted tissues in 4 dimensions (3D plus time) in real time using existing technologies such as multiphoton microscopy. We transplanted a variety of C57BL/6 adult (lung, trachea, aorta, kidney, bone marrow, thymus, spleen, lymph node, skeletal muscle, thyroid gland, adrenal gland) and fetal (colon, ileum, stomach, heart, lung, kidney, bone marrow, thymus, spleen, skeletal muscle) tissues subcutaneously into syngeneic mouse ear pinna. All of these tissues were able to survive at least 4–8 weeks after transplantation. Many of these tissues maintained normal or close to normal structures for at least 4–8 weeks. We chose an ear-thymus model to test whether the engrafted tissues can function. Thymic tissue from C57BL/6 newborn mice (<48 hrs) was transplanted into BALB/c nude mice (lacking thymic tissue). The numbers of CD4+ and CD8+ T cells were followed by flow cytometric analysis in peripheral blood over time. CD4+ T cell counts were significantly higher in thymic tissue recipients compared with sham transplanted control group (Figure, P<0.05). As an internal control, B220+ B cells, which are normal in nude mice and are not produced in thymus, remained similar between transplanted and sham transplanted groups at all time points. To test whether this model is useful for high-resolution imaging in live animals, we transplanted GFP+ C57BL/6 T cell depleted bone marrow into thymic tissue recipients. After bone marrow transplantation, almost all hematopoietic cells were replaced by GFP+ cells. Using two photon microscopy technology, we were able to obtain 4 dimension images of the transplanted thymic tissue at the cellular level in living animals. Because surgical exposure is not required, we were able to perform imaging of living tissues repeatedly in these animals indefinitely. We conclude that multiple tissues are able to survive and function for a long period of time when transplanted into ear pinna. Our innovative ear-tissue transplant model has the potential to allow many living tissues to be visualized at the cellular and subcelluar level in real time and in live animals. Disclosures: Chao: Genzyme: Research Funding.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1732-1732
Author(s):  
Suigui Wan ◽  
Changqing Xia ◽  
Laurence Morel

Abstract The B6.Sle1.Sle2.Sle3 triple congenic mouse (B6.TC) is a model of lupus due to the co-expression of the three major NZM2410-derived susceptibility loci on a C57BL/6 background. B6.TC mice produce high titers of anti-nuclear nephrogenic autoantibodies and a highly penetrant glomerulonephritis. Previous studies have shown the Sle1 locus is associated with a reduced number of regulatory T cells (Treg), and that Sle3 results in intrinsic defects in myeloid cells that hyperactivate T cells. Here, we show that B6.TC dendritic cells (DCs) accumulate in lymphoid organs and present a defective maturation process, in which bone-marrow derived DCs, plasmacytoid and myeloid DCs express a significantly lower level of CD80, CD86 and class II MHC than B6 controls. B6.TC DCs also induce a higher level of proliferation in CD4+ T cells than B6 DCs, and B6.TC DCs block the suppressive activity of Treg. B6.TC DCs over-produce IL-6, which is necessary for the blockade of Treg activity, as shown by anti-IL-6 neutralizing antibody in the suppression assays. The over-production of IL-6 by DCs and the blockade of Treg activity maps to Sle1, which therefore not only confers a reduced number of Treg, but also blocks their ability to regulate autoreactive T cells. Taken together, these results provide a genetic and mechanistic evidence for systemic autoimmunity resulting from an impaired regulatory T cell compartment both in number and function, and for Sle1-expressing DCs playing a major role in the latter defect though their production of IL-6.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1116-1116
Author(s):  
Jeong-Su Do ◽  
Alex Y. Huang ◽  
Daniel Zwick ◽  
Fei Zhong ◽  
David Askew ◽  
...  

Abstract Tumor growth factor β (TGF-β)-induced peripheral regulatory T cells (pTreg) are a promising therapeutic cell source that exhibit Foxp3 expression and suppressive functions similar to natural regulatory T cells. Nonetheless, their clinical potential is limited by the instability of Foxp3 expression and T cell exhaustion that occurs during ex vivo expansion. We postulated that mesenchymal stromal cells (MSCs) could enhance the number, function and Foxp3 expression stability of pTregs during IL-2 driven 21 day expansion due to their diverse immunomodulatory properties. In this study, we observed that use of a human bone marrow mesenchymal stromal cells (hBM-MSC) platform significantly enhanced the number of pTreg during IL-2 driven 21 day ex vivo expansion vs. standard suspension culture condition (MSC platform: 80.2 x 106 vs. IL2/media: 39.3 x 106, n=6; p<0.01). Also the number of pTreg expressing a naive phenotype (CD4+CD45RA+ and CD4+CD62L+ ) were significantly increased (CD45RA+; MSC platform: 74.4 ± 1.6 x 106 vs. IL2/media: 45.9 ± 2.9 x 106, n=6, p<0.001; CD62L+; MSC platform: 79.1 ± 1.3 x 106 vs. IL2/media: 54.5 ± 2.1 x 106, n=6, p<0.001), as well as stability of Foxp3 expression (IL-2/media: 88.2 ± 1.7% vs. MSC platform: 96.2 ± 1.1%, n=7; p<0.05). In addition, pTreg suppressive function was noted to be more potent during 21 day IL-2 driven ex vivo expansion compared to standard IL-2/media culture condition (MSC platform: 79% vs. media: 35% inhibition of T cell proliferation in 10:1 ratio, n=6; p<0.01). pTreg expanded over a hBM-MSC platform exhibited higher surface CD25, CTLA-4, and ICOS MFI expression (CD25; MSC platform: 1410 vs. Media: 774; p<0.001, CTLA-4; MSC platform: 1084 vs. Media: 318; p<0.001, ICOS; MSC platform: 4386 vs. Media: 2641, p<0.01, n=6). Notably, hBM-MSC enhancement of pTreg ex vivo expansion requires direct cell-cell contact, as Foxp3 expression in pTreg was not enhanced by hBM-MSC conditioned media (CM:73.4 ± 6.8% vs. MSC platform: 96.2 ± 1.0%, p<0.001; and IL2/media: 88.8 ± 1.6% vs. MSC platform: 96.2 ± 1.0%, p<0.01) nor in a trans-well culture experiments (Transwell: 83.4 ± 2.5% vs. IL2/media: 88.8 ± 1.6%; and Transwell: 83.4 ± 2.5% vs. MSC platform: 96.2 ± 1.0%, p<0.01). Importantly, optical sectioning microscopy and flow cytometry revealed that hBM-MSC supports Treg number and function via direct contact-dependent mitochondrial transfer (Figure 1A-B). Cytochalasin B treatment blocked mitochondrial transfer, suggesting that tunneling nanotubes (TNT) facilitate mitochondrial transfer from hBM-MSC to pTreg during IL-2 driven ex vivo expansion (Mock: 2208 ± 122.1 vs. Cyto B: 923.8 ± 89 MFI, n=6, p<0.0001). Moreover, the quantity of ATP (n=6; p<0.01) mitochondrial potential of pTreg (MSC platform: 9010 ± 224.5 vs. media: 7316 ± 122.7 MFI, n=6; p<0.01) were significantly enhanced in pTreg during IL-2 driven ex vivo expansion over a hBM-MSC platform. Taken together, hBM-MSC significantly improves the number, maturation, and function of pTreg during 21 day IL-2 driven ex vivo expansion. We have identified one key mechanism of action of hBM-MSC underlying these favorable effects on pTreg during ex vivo expansion to be mitochondrial transfer via TNT. Notably, these studies identify a novel role of hBM-MSC to overcome current limitations in IL-2/media suspension culture conditions including T cell senescence, and loss of Foxp3 expression. Disclosures No relevant conflicts of interest to declare.


2015 ◽  
Vol 2015 ◽  
pp. 1-12 ◽  
Author(s):  
Minhwa Park ◽  
Yu-Hee Kim ◽  
Jung-Hwa Ryu ◽  
So-Youn Woo ◽  
Kyung-Ha Ryu

Mesenchymal stem cells (MSCs) are considered valuable sources for cell therapy because of their immune regulatory function. Here, we investigated the effects of tonsil-derived MSCs (T-MSCs) on the differentiation, maturation, and function of dendritic cells (DCs). We examined the effect of T-MSCs on differentiation and maturation of bone-marrow- (BM-) derived monocytes into DCs and we found suppressive effect of T-MSCs on DCs via direct contact as well as soluble mediators. Moreover, T cell proliferation, normally increased in the presence of DCs, was inhibited by T-MSCs. Differentiation of CD4+T cell subsets by the DC-T cell interaction also was inhibited by T-MSCs. The soluble mediators suppressed by T-MSCs were granulocyte-macrophage colony-stimulating factor (GM-CSF), RANTES, interleukin-6 (IL-6), and monocyte chemoattractant protein-1 (MCP-1). Taken together, T-MSCs exert immune modulatory function via suppression of the differentiation, maturation, and function of BM-derived DCs. Our data suggests that T-MSCs could be used as a novel source of stem cell therapy as immune modulators.


2011 ◽  
Vol 91 (2) ◽  
pp. 197-205 ◽  
Author(s):  
Dietmar Herndler-Brandstetter ◽  
Katja Landgraf ◽  
Alexandar Tzankov ◽  
Brigitte Jenewein ◽  
Regina Brunauer ◽  
...  

Author(s):  
Jin Yan Yap ◽  
Leen Moens ◽  
Ming-Wei Lin ◽  
Alisa Kane ◽  
Anthony Kelleher ◽  
...  

Abstract Purpose Deficiency of adenosine deaminase type 2 (ADA2) (DADA2) is a rare inborn error of immunity caused by deleterious biallelic mutations in ADA2. Clinical manifestations are diverse, ranging from severe vasculopathy with lacunar strokes to immunodeficiency with viral infections, hypogammaglobulinemia and bone marrow failure. Limited data are available on the phenotype and function of leukocytes from DADA2 patients. The aim of this study was to perform in-depth immunophenotyping and functional analysis of the impact of DADA2 on human lymphocytes. Methods In-depth immunophenotyping and functional analyses were performed on ten patients with confirmed DADA2 and compared to heterozygous carriers of pathogenic ADA2 mutations and normal healthy controls. Results The median age of the patients was 10 years (mean 20.7 years, range 1–44 years). Four out of ten patients were on treatment with steroids and/or etanercept or other immunosuppressives. We confirmed a defect in terminal B cell differentiation in DADA2 and reveal a block in B cell development in the bone marrow at the pro-B to pre-B cell stage. We also show impaired differentiation of CD4+ and CD8+ memory T cells, accelerated exhaustion/senescence, and impaired survival and granzyme production by ADA2 deficient CD8+ T cells. Unconventional T cells (i.e. iNKT, MAIT, Vδ2+ γδT) were diminished whereas pro-inflammatory monocytes and CD56bright immature NK cells were increased. Expression of the IFN-induced lectin SIGLEC1 was increased on all monocyte subsets in DADA2 patients compared to healthy donors. Interestingly, the phenotype and function of lymphocytes from healthy heterozygous carriers were often intermediate to that of healthy donors and ADA2-deficient patients. Conclusion Extended immunophenotyping in DADA2 patients shows a complex immunophenotype. Our findings provide insight into the cellular mechanisms underlying some of the complex and heterogenous clinical features of DADA2. More research is needed to design targeted therapy to prevent viral infections in these patients with excessive inflammation as the overarching phenotype.


1979 ◽  
Vol 150 (4) ◽  
pp. 808-817 ◽  
Author(s):  
A F Schrater ◽  
E A Goidl ◽  
G J Thorbecke ◽  
G W Siskind

Although athymic mice make an excellent immune response to the thymus-independent antigen trinitrophenyl-lys-Ficoll (TNP-F), nude mice of AKR/J and BALB/c strains lack the anti-idiotypic response that occurs in euthymic mice of both of these strains within the first 1--2 wk after injection of more TNP-F. Anti-idiotypic antibody-blocked (hapten-augmentable) anti-TNP splenic plaque-forming cells (PFC) do not occur at any time and serum anti-idiotypic antibody is absent in both congenitally athymic mice, and thymectomized, irradiated, bone marrow-reconstituted mice. Nevertheless, nu/nu mice do have PFC which can be inhibited by exposure to anti-idiotypic antibody produced in +/+ mice. As a consequence of the failure to produce anti-idiotypic antibodies, the anti-TNP PFC response is athymic as compared to euthymic mice is of greater magnitude, declines less precipitously, and shows an increase rather than a decrease in affinity between days 4 and 7 after antigen injection. It is concluded that the anti-idiotypic antibody response is thymus dependent and that athymic mice lack a helper cell required for the induction of anti-idiotypic antibodies.


Blood ◽  
2014 ◽  
Vol 124 (6) ◽  
pp. 963-972 ◽  
Author(s):  
Angela Mensen ◽  
Korinna Jöhrens ◽  
Ioannis Anagnostopoulos ◽  
Sonya Demski ◽  
Maike Oey ◽  
...  

Key Points Donor T-cell infiltration of the bone marrow is associated with impaired B-cell immunity after allogeneic HSCT. Quantification of κ-deleting recombination excision circles as a biomarker for bone marrow B-cell output in different clinical episodes.


1989 ◽  
Vol 24 (2) ◽  
pp. 111-124
Author(s):  
Brigitte Autran ◽  
Guy Gorochov ◽  
Ioannis Theodorou ◽  
Marie Claude Couty ◽  
Patrice Debre

Sign in / Sign up

Export Citation Format

Share Document