In Vivo Bioluminescence Imaging to Study the Contribution of TNF-TNFR Interactions on Immune and Parenchymal Cells to Tumor Cell Progression in a Syngenic Mouse Model

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1110-1110
Author(s):  
Martin Chopra ◽  
Simone S Riedel ◽  
Viktoria von Krosigk ◽  
Carina A Bäuerlein ◽  
Christian Brede ◽  
...  

Abstract Abstract 1110 The cytokine tumor necrosis factor-α (TNF) has pleiotropic functions both in normal physiology and disease. TNF and its relative lymphotoxin-α (LT) signal by activating two cell surface receptors TNFR1 and TNFR2. TNFR1 is expressed on most cells whereas TNFR2 is mainly expressed in cells of the hematopoietic system. TNF-TNFR interactions were shown to play a major role in graft-versus-leukemia effect and in the immunosurveillance of solid tumors. To study the contribution of TNF-TNFR interactions on tumor cell progression we employed a syngenic B16 melanoma mouse model combined with in vivo bioluminescence imaging. Firefly luciferase-transgenic B16 melanoma cells were injected intravenously into syngenic albino C57BL/6 hosts. The host mice were either of wildtype, TNF, LT, TNFR1, TNFR2 knockout or TNFR1R2 double knockout genotype. The localization and expansion of the B16 cells was monitored by in vivo bioluminescence imaging for up to 14 days. On days 15, mice were sacrificed and internal organs were imaged ex vivo to further elucidate the organ-specific tumor burden. B16 tumors were primarily found in the lungs of all genotypes. All female knockout genotypes displayed a higher lung tumor burden than wildtype mice. In male mice, only TNF knockout presented enhanced tumor cell signals. Following ex vivo imaging we evaluated the pulmonary infiltration of NK1.1 or NKp46, CD8, CD4 and CD4/CD25/Foxp3 regulatory T cells by flow cytometry and immunofluorescence microscopy. Compared to wildtype mice, more regulatory T cells infiltrated the lungs of female TNFR1 knockout mice (200%). In LT knockout mice, very few NK cells (<20%) but more CD4+ cells (160%) infiltrated the lungs. Only subtle changes occurred in the other deficient mouse strains. However, these changes were independent of the presence of tumor cells and could also be found in normal knockout mice without B16 tumors. Within sections of tumor-bearing lungs, we found that TNF and all three TNFR knockouts exhibited less CD8+ cells within tumors than did wildtype or LT knockout mice. The number of CD8+ cells in normal lung tissue was not altered across the different genotypes. The deficit in NK cells of LT knockout mice was confirmed by histology. The enhanced tumor progression in all knockout mice could be a secondary effect due to their altered immune phenotype rather than to the loss of TNF-TNFR interactions. To circumvent this potential experimental bias and to further assess the influence of the loss of expression of parts of the TNF/TNFR-system in immune cells only, we generated bone marrow chimeras by reconstituting lethally irradiated female wildtype mice with bone marrow derived from TNF, LT, TNFR1 or TNFR2 knockout mice. Tumor cell signals in these chimeric mice progressed more than in normal wildtype mice. In contrast to the first set of experiments with knockout mice, we found that mice reconstituted with either TNF or TNFR2 knockout bone marrow presented less tumor cell signal than did mice reconstituted with wildtype bone marrow. TNF-TNFR interactions between immune cells appear to exhibit pro-tumorigenic functions in our mouse model. These results show that TNF-TNFR interactions are an important step in tumor cell progression and that the outcome of these interactions differs, depending on whether immune or parenchymal cells are deficient in TNF-TNFR signalling components. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3066-3066
Author(s):  
Michael P. Rettig ◽  
Julie K. Ritchey ◽  
Julie L. Prior ◽  
David Piwnica-Worms ◽  
Mark L. Bonyhadi ◽  
...  

Abstract Herpes simplex virus thymidine kinase (TK) gene-modified T cells are currently being evaluated in gene therapy clinical trials for the control of graft-versus-host disease (GVHD) after allogeneic BMT. Unfortunately, these trials have been limited by a consistent failure of the ex-vivo manipulated T cells to survive and function properly in vivo. We recently developed a technique for retrovirally transducing and selecting murine T cells with a novel chimeric CD34-TK fusion suicide gene that preserves their alloreactivity after allogeneic BMT. In this study, we assessed the trafficking, survival, and GVHD-inducing potential of ex vivo manipulated murine T-cells in fully allogeneic transplant recipients by in vivo bioluminescence imaging (BLI) with two novel reporter vectors. The first vector encodes a fusion protein comprised of click beetle red (CBR) luciferase and EGFP (CBR/EGFP). In the second vector, we inserted a click beetle green (CBG) luciferase between CD34 and TK in our chimeric suicide gene (CD34/CBG/TK). Murine T cells, stimulated 24 h with anti-CD3 and anti-CD28 antibody-coated magnetic beads (CD3/CD28 beads), were transduced with Phoenix-Eco-derived CBR/EGFP or CD34/CBG/TK retrovirus and purified to &gt;85% using a MoFlo cell sorter or CD34 immunomagnetic selection 48 h post-infection. To induce GVHD, lethally irradiated BALB/c allogeneic recipients were given T cell depleted C57BL/6 (B6) bone marrow supplemented with either 1e6 CBR/EGFP or CD34/CBG/TK purified B6 T cells. The CBR/EGFP BLI signal was significantly increased over background at 24 h post-injection, with the allogeneic T cells localizing primarily to the spleen and secondary lymph nodes. Over the next 2–3 days the CBR/EGFP+ cells migrated to the entire intestinal area followed rapidly by infiltration of the skin. Overall, the CBR/EGFP BLI signal increased nearly 3 orders of magnitude between days 1 and 8 post-BMT, remained steady for a week, and then only gradually declined over the next month (only a 3-fold decrease between days 14 to 42 post-BMT). Consistent with GVHD, these mice lost &gt;20% of their pretransplant body weight and exhibited impaired lymphoid reconstitution. We observed similar trafficking and GVHD-inducing potential when CD34/CBG/TK gene-modified T cells were injected into BALB/c recipients. However, the maximum BLI signal intensity from the CD34/CBG/TK T cells was decreased nearly 2 orders of magnitude compared to the CBR/EGFP-modified T cells. Nevertheless, we were still able to demonstrate a significant reduction in BLI signal intensity when recipients of CD34/CBG/TK-modified allogeneic T cells were treated with ganciclovir (GCV) from days 1 to 7 post-BMT. This observation is consistent with in vitro cell sensitivity assays, which demonstrated that cells modified with the CD34/CBG/TK reporter gene retain TK activity similar to CD34-TK modified cells. In summary, this study demonstrated by in vivo BLI that allogeneic murine T cells activated and expanded ex vivo with CD3/CD28 beads retain significant GVHD-inducing potential and can be eliminated by HSV-TK/GCV suicide gene therapy.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1809-1809
Author(s):  
Sabrina Kraus ◽  
Martin Chopra ◽  
Christian Brede ◽  
Simone S Riedel ◽  
Mike Friedrich ◽  
...  

Abstract Abstract 1809 Various imaging platforms are well established in hematology research. Nevertheless, the three-dimensional architecture of the bone marrow and tumor growth within this microenvironment remain largely uncharacterized. To date the major hindrance to microscopically image tumor engraftment and the immune response in the bone marrow on a single cell level is the compact structure of the bone that is almost impossible to image through. Therefore, we developed a novel bioluminescent mouse model that recapitulates the clinical characteristics of MM using the new human UMM3 cell line (CD38+, CD56+, CD138+, CD19−, CD20−), from the pleural effusion of a patient with an IgG lambda myeloma (ISS stage I) as well as the well-characterized RPMI8226 cell line transduced to express eGFP and firefly luciferase (UMM3eGFPluc and RPMIeGFPluc). 1×106 MM cells were injected intravenously into NOD.Cg-Prkdcscid IL2rg (NSG) mice and disease progression and bone marrow (BM) engraftment were monitored twice weekly by in vivo bioluminescence imaging. Both cell lines homed to the BM compartment, reflecting MM pathophysiology. Histological analysis confirmed BM engraftment and showed multiple osteolytic lesions for both UMM3 and RPMI cells. Since we were interested in imaging the interactions between human MM cells and the bone marrow microenvironment on a single cell level, we employed the multi-color LSFM after decalcification, specific deep-tissue antibody staining and clearing of the bone structures. With this innovative microscopy technique, we were able to establish a novel tool to display tumor cell engraftment in the bone marrow compartment in three dimensions through the intact bone. We recorded 1500 optical sections for three individual channels each (488, 532, and 647 nm) with an increment of 5μm which allows scanning the whole bone marrow compartment of the sternum within minutes in single-cell resolution. Using higher magnification enabled us to even visualize subcellular components within the bone marrow. Moreover, tissue autofluorescene, recorded mainly in the 488 nm channel, displayed detailed microanatomical structures which allowed for the localization of individual cells within their anatomical context. We could establish protocols for various fluorophore-coupled antibodies and successfully stained CD138+ cells in relation to CD3+ cells and to the microenvironment in the bone marrow. The CD138-positive cells infiltrated the bone marrow in a number of small clusters and comprising about 15% of cellular elements in total. Ex vivo bioluminescence imaging of the sternum from UMM3 tumor-bearing mice revealed massive infiltration of luciferase-expressing cells into the bone marrow compartment. This could also be confirmed by flow cytometrical analysis of bone marrow cells which showed eGFP+hCD138+ cells. We have successfully introduced a novel technique to study MM cell engraftment and progression in a humanized mouse model. We were able to track the tumor cells both in the living animal by in vivo bioluminescence imaging and on single-cell resolution by multi-color LSFM within the intact bone. Our model may lead to better insights into the pathogenesis of MM and could serve as a model for preclinical testing of new therapeutic approaches for the treatment of MM patients. Disclosures: No relevant conflicts of interest to declare.


PLoS ONE ◽  
2015 ◽  
Vol 10 (3) ◽  
pp. e0120925 ◽  
Author(s):  
Binje Vick ◽  
Maja Rothenberg ◽  
Nadine Sandhöfer ◽  
Michela Carlet ◽  
Cornelia Finkenzeller ◽  
...  

2004 ◽  
Vol 120 (2) ◽  
pp. 249-255 ◽  
Author(s):  
Shiva Sarraf-Yazdi ◽  
Jing Mi ◽  
Mark W. Dewhirst ◽  
Bryan M. Clary

2011 ◽  
Vol 30 (3) ◽  
pp. 335-340 ◽  
Author(s):  
Jonathan R. Pribaz ◽  
Nicholas M. Bernthal ◽  
Fabrizio Billi ◽  
John S. Cho ◽  
Romela Irene Ramos ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1834-1834 ◽  
Author(s):  
Edmund Lee ◽  
Bret Bannerman ◽  
Michael Fitzgerald ◽  
Jennifer Terkelsen ◽  
Daniel Bradley ◽  
...  

Abstract Abstract 1834 Poster Board I-860 Introduction The clinical success of VELCADE® (bortezomib) for Injection has validated the proteasome as a therapeutic target for the treatment of human cancer. The novel proteasome inhibitor MLN9708 is a potent, reversible, and specific inhibitor of the b5 site of the 20S proteasome identified in preclinical studies. MLN9708 is currently in human clinical development for both hematological and non-hematological malignancies. Here we describe the pharmacodynamic (PD) response of MLN9708 in the murine bone marrow compartment and its strong antitumor activity in an intraosseous xenograft model of plasma cell malignancy. Materials MLN9708 immediately hydrolyzes to MLN2238, the biologically active form, upon exposure to aqueous solutions or plasma. MLN2238 was used for all preclinical studies described below. Methods It has been previously shown that double transgenic iMycCa/Bcl-XL mice develop de novo plasma cell malignancies (J. Clin. Invest. 113:1763-1773, 2004) in which neoplastic plasma cell development is driven by the targeted expression of the transgene Myc (c-myc; myelocytomatosis oncogene) and Bcl-x (Bcl2l1; encodes the oncoprotein Bcl-XL). DP54 is a plasma cell tumor cell line derived from the bone marrow of a syngeneic mouse previously inoculated with an iMycCa/Bcl-XL tumor (Cancer Res. 67:4069-4078, 2007). In vitro, DP54 cells express both the Myc and Bcl-XL transgenes, various plasma cell and B-cell markers including CD38, CD138 and B220, and has gene expression profile very similar to human multiple myeloma. To establish a preclinical intraosseous model of plasma cell malignancy for efficacy studies, freshly dissociated DP54-Luc cells (constitutively expressing firefly luciferase under a mouse Ig-k promoter) were aseptically injected into the bone marrow space of the upper shaft of the right tibia of NOD-SCID mice. Once tumor growth has been established, mice were randomized into treatment groups and then treated intravenously (IV) with vehicle, bortezomib (at 0.8 mg/kg twice weekly [BIW]) or MLN2238 (at 11 mg/kg BIW) for 3 consecutive weeks. Tumor burden was measured by bioluminescent imaging. Results MLN2238 strongly inhibited proteasome activity in the blood and bone marrow compartments of mice (maximum b5 inhibition of 84% and 83%, respectively). In vivo, when DP54 cells were aseptically injected into the bone marrow space of the mouse tibia, signs of bone erosion in the tibia, femur and cranial sagittal sultures (as determined by ex-vivo mCT imaging) were observed which resembled osteolytic lesions frequently seen in human multiple myeloma. Dissemination of DP54-Luc cells after intratibia inoculations were detected by in vivo bioluminescent and confirmed by ex vivo imaging where luminescent tumor nodules were detected in the spleen, kidneys, intestine, lymph nodes and bones including right tibia, spine and cranium. To assess the antitumor activity of MLN2238 in the bone marrow compartment, an efficacy study was performed using the DP54-Luc intraosseous xenograft model of plasma cell malignancy. Tumor burden (bioluminescence), osteolytic lesions (mCT) and overall survival after treatment with bortezomib and MLN2238 will be presented. Conclusion The novel proteasome inhibitor MLN9708 demonstrates strong activity in the bone marrow compartment in vivo. MLN9708 is currently in human clinical development for both hematological and solid tumor indications. Disclosures Lee: Milllennium: Employment, Equity Ownership. Bannerman:Milllennium: Employment. Terkelsen:Milllennium: Employment. Bradley:Milllennium: Employment, Equity Ownership, Research Funding. Li:Milllennium: Employment. Li:Milllennium: Employment. Janz:Milllennium: Research Funding. Van Ness:Milllennium: Research Funding. Manfredi:Milllennium: Employment. Kupperman:Milllennium: Employment.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Jing Zhang ◽  
Yaozhen Chen ◽  
Dandan Yin ◽  
Fan Feng ◽  
Qunxing An ◽  
...  

Abstract Background The fate of hematopoietic stem cells (HSCs) is determined by a complex regulatory network that includes both intrinsic and extrinsic signals. In the past decades, many intrinsic key molecules of HSCs have been shown to control hematopoiesis homeostasis. Non-hematopoietic niche cells also contribute to the self-renewal, quiescence, and differentiation of HSCs. Mesenchymal stromal cells (MSCs) have been identified as important components of the niche. However, the regulatory role of MSCs in hematopoiesis has not been fully understood. Methods Caspase-3 and NLRP3 gene knockout mice were generated respectively, and hematopoietic development was evaluated in the peripheral circulation and bone marrow by flow cytometry, colony formation assay, and bone marrow transplantation. Bone-associated MSCs (BA-MSCs) were then isolated from gene knockout mice, and the effect of Caspase-3/NLRP3 deficient BA-MSCs on hematopoiesis regulation was explored in vivo and ex vivo. Results We report that Caspase-3 deficient mice exhibit increased myelopoiesis and an aberrant HSC pool. Ablation of Caspase-3 in BA-MSCs regulates myeloid lineage expansion by altering the expression of hematopoietic retention cytokines, including SCF and CXCL12. Interestingly, NLRP3 gene knockout mice share phenotypic similarities with Caspase-3 deficient mice. Additionally, we found that NLRP3 may play a role in myeloid development by affecting the cell cycle and apoptosis of hematopoietic progenitors. Conclusions Our data demonstrate that the Caspase-3/NLRP3 signaling functions as an important regulator in physiological hematopoiesis, which provides new insights regarding niche signals that influence hematopoiesis regulation in the bone marrow.


2016 ◽  
Vol 2016 ◽  
pp. 1-11 ◽  
Author(s):  
Juan Cao ◽  
Shike Hou ◽  
Hui Ding ◽  
Ziquan Liu ◽  
Meijuan Song ◽  
...  

Recently, mesenchymal stem cells (MSCs) are increasingly used as a panacea for multiple types of disease short of effective treatment. Dozens of clinical trials published demonstrated strikingly positive therapeutic effects of MSCs. However, as a specific agent, little research has focused on the dynamic distribution of MSCs afterin vivoadministration. In this study, we track systemically transplanted allogeneic bone marrow mesenchymal stem cells (BMSCs) in normal rats through bioluminescence imaging (BLI) in real time.Ex vivoorgan imaging, immunohistochemistry (IHC), and RT-PCR were conducted to verify the histological distribution of BMSCs. Our results showed that BMSCs home to the dorsal skin apart from the lungs and kidneys after tail vein injection and could not be detected 14 days later. Allogeneic BMSCs mainly appeared not at the parenchymatous organs but at the subepidermal connective tissue and adipose tissue in healthy rats. There were no significant MSCs-related adverse effects except for transient decrease in neutrophils. These findings will provide experimental evidences for a better understanding of the biocharacteristics of BMSCs.


Sign in / Sign up

Export Citation Format

Share Document