A New In Vivo Platform for Studying the Growth of Hematopoietic Cancer Initiating Blood Cells and Response to Therapy.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4142-4142
Author(s):  
Michal Cipok ◽  
Yona Farnoushi ◽  
Sigi Kay ◽  
Igor Grinberg ◽  
Arbel Reis ◽  
...  

Abstract Abstract 4142 There are few in-vivo models for studying human leukemia and its therapy. These include the high cost immune-deficient NOD/SCID mice and large mammalian fetuses, which both require weeks to assess treatment response. We described a rapid and low-cost alternative in-vivo system for human leukemia in the preimmune chick embryo (Taizi et al, Exp Hem 34;1698-708,2006). We recently demonstrated that the turkey embryo provides a more robust model for the preclinical assessment of human leukemia infiltration (Grinberg et al, Leukemia Res 33;1417-26 2009). Here we describe the application of this powerful and inexpensive model for rapid preclinical assessment of anti-cancer therapies and basic research of blood malignancies. BM engraftment was robust occurring in 95% of the embryos using cells lines and 40% using fresh samples. Leukemia cells homed to the BM where they were already detected at 20 hours after injection and reached highest levels on days E19-24, using FACS and RT-PCR. Serial engraftment in secondary recipients of all three human stem cell leukemia lines and one fresh sample was detected in embryos injected with BM harvested 8-10 days after the first inoculation, validating the engraftment of cancer initiating cells. Human stem cell leukemia lines K562 and LAMA, (both BCR/Abl+) and CHRF (c-Kit+), and myeloma cell lines ARH-77 and CAG and fresh patient samples were injected IV into turkeys, on embryonic day E 11, using 5×106 cells from lines or 107 fresh patient cells. Engraftment of human leukemia and myeloma cells (cell lines and fresh samples) was detected 8-14 days later (E19-24), in the BM and in several hematopoietic organs at a frequency of 0.5->20%, by real-time PCR, immunohistochemistry and flow cytometry. CAG and ARH-77 myeloma cells engraftment was also detected by the presence of human monoclonal free light chain (6-10 mg/L) in blood collected from vessels of the chorioallantoic membrane, one week after cell injection. The growth of leukemias treated with doxorubicin or the tyrosine-kinase inhibitor Imatinib and myeloma with Velcade, at levels that were not toxic to the developing embryonic BM, was dramatically inhibited in vivo when the drug was injected together with cells on E11 or 48-72 hours after injection of the cells and homing to the BM. Using flow cytometry analysis the frequency of CHRF cells (detected with anti-human CD33) was reduced from 8% to 0.01% and K562 and LAMA (detected with anti-human CD71) from 1%-3% engraftment to <0.17% following treatment with Imatinib. Q-PCR analysis supported these results showing an average 8 fold reduction of CHRF and a 2-5 fold reduction of K562 and LAMA cells in the Imatinib treated turkey embryos. The ARH-77 cells (detected with anti-human CD38 and CD138) were inhibited from 8.5% to 0.72% after Velcade treatment, with a 16.5 fold reduction determined by Q-PCR analysis compare to untreated embryos. These results prove the efficacy and demonstrate the utility of the turkey embryo as a new complementary in-vivo model for studying cancer initiating cells and the growth of human blood malignancies and their response to treatment. With further improvements, it may provide an affordable, rapid in vivo system for studying the growth blood malignancies and help reduce time and cost of drug development. Disclosures: No relevant conflicts of interest to declare.

2020 ◽  
Vol 15 (1) ◽  
pp. 83-91
Author(s):  
Chen Li ◽  
Biao Qian ◽  
Zhao Ni ◽  
Qinzhang Wang ◽  
Zixiong Wang ◽  
...  

AbstractThis study aims to construct recombinant lentiviral vectors containing the human stem cell leukemia (SCL) gene and investigate their in vitro transfection efficiency in Interstitial Cells of Cajal (ICC) of guinea pig bladders. In this study, the human SCL gene was successfully cloned, and the recombinant lentivirus GV287-SCL was successfully constructed. The titer of the recombinant lentivirus was 5 × 108 TU /mL. After transfecting the ICCs with the lentiviral vector at different MOIs, the optimal MOI was determined to be 10.0, and the optimal transfection time was determined to be 3 days. The amplification product of the lentivirus transfection group was consistent with the target fragment, indicating that the SCL gene had been successfully introduced into ICCs. In conclusion, the recombinant lentiviral vector GV287-SCL was successfully constructed and transfected into the in vitro cultured ICCs. The successful expression of SCL in ICCs may provide an experimental basis for the in vivo transfection of the SCL gene.


1989 ◽  
Vol 170 (1) ◽  
pp. 339-342 ◽  
Author(s):  
C G Begley ◽  
P D Aplan ◽  
M P Davey ◽  
J P de Villartay ◽  
D I Cohen ◽  
...  

It has been hypothesized that a rearrangement between the delta recombining element (delta Rec) and a pseudo J alpha gene serves to delete the TCR-delta locus before rearrangement of the TCR-alpha genes. We have now sequenced a direct, site-specific rearrangement between the delta Rec element and a pseudo J alpha gene in a human leukemic stem-cell line. Putative "N-sequence" addition was noted at the site of recombination, suggesting that this event occurred at a time when the enzyme(s) involved in N-region addition were active in this cell. This provides support for the view that deletion of the TCR-delta locus is required before rearrangement of the TCR-alpha chain genes.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2549-2549
Author(s):  
Jing Chen ◽  
Daniel J. DeAngelo ◽  
Jeffery L. Kutok ◽  
Ifor R. Williams ◽  
Benjamin H. Lee ◽  
...  

Abstract Human stem cell leukemia-lymphoma syndrome usually presents as a myeloproliferative disease (MPD) that evolves to acute myeloid leukemia and/or lymphoma. The syndrome associated with t(8;13)(p11;q12) results in expression of the ZNF198-FGFR1 fusion tyrosine kinase. Current empirically-derived cytotoxic chemotherapy is inadequate treatment of this disease. We hypothesized that small molecule inhibitors of the ZNF198-FGFR1 fusion would have therapeutic efficacy. We characterized the transforming activity of ZNF198-FGFR1 in hematopoietic cells in vitro and in vivo. Expression of ZNF198-FGFR1 in primary murine hematopoietic cells caused a myeloproliferative syndrome in mice that recapitulated the human MPD phenotype. Transformation in these assays, and activation of the downstream effector molecules PLCγ, STAT5 and PI3K/AKT, required the proline-rich, but not the zinc-finger domains of ZNF198. A small molecule tyrosine kinase inhibitor, PKC412 (N-benzoyl-staurosporine) effectively inhibited ZNF198-FGFR1 tyrosine kinase activity and activation of downstream effector pathways, and inhibited proliferation of ZNF198-FGFR1 transformed Ba/F3 cells. Furthermore, treatment with PKC412 resulted in statistically significant prolongation of survival in the murine model of ZNF198-FGFR1 induced myeloproliferative disease. Based in part on these data, PKC412 was administered to a patient with t(8;13)(p11;q12) and was efficacious in treatment of progressive myeloproliferative disease with organomegaly. Therefore, PKC412 may be a useful therapy for treatment of human stem cell leukemia-lymphoma syndrome.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4073-4073
Author(s):  
Varda Deutsch ◽  
Yona Farnoushi ◽  
Michal Cipok ◽  
Sigi Kay ◽  
Elizabeth Naparstek ◽  
...  

Abstract Abstract 4073 While new treatment options are available, multiple myeloma (MM) still remains an incurable malignancy of plasma cells with a grim prognosis. Practical in vivo models to study human MM may enable a better understanding of the biology of the disease, and better optimization of therapeutic strategies. The best current xenograft model, the immune-deficient NOD/SCID mice, recapitulates MM in vivo, however, the price is very costly and maintenance complex, with >1 month required to establish engraftment. Our goal was to develop a user friendly rapid alternative xenograft system for the preclinical assessment of MM growth and therapy. We recently described this new in-vivo system for studying human leukemia in the pre-immune turkey embryo 1,2. These embryos are inexpensive, require no maintenance, and are easily manipulated experimentally. Described here are the first attempts at application of this novel system to study MM and test therapies. Cell lines ARH-77 and CAG line and fresh patient cells (5 × 106/embryo) were injected IV into turkey egg chorioallantoic membrane veins on embryonic day E11. Engraftment of human cells in hematopoietic organs, bone marrow (BM) and liver was detected 7 days later (E18) by RTPCR, immunohistochemistry and flow cytometry and by circulating free light chain (6-25 mg/L) in the peripheral blood of 100% of the injected cell lines and 50% of patients myelomas. Treatment with Velcade (Bortezomib) or Revlimid IV on E13 (48 hours after MM cell injection), at drug levels that were precalibrated to be non-toxic to the developing embryonic BM, dramatically reduced engraftment, demonstrating the utility of this new model for testing drug activity in vivo. ARH-77 cells, detected by flow cytometry of the embryonic BM cells with anti-human CD19, CD38 and CD138, were inhibited from 8.5% engraftment to 0.72% after a single Velcade treatment, with an 18 fold decrease compared to untreated embryos in the ratio of human to avian cells in BM tissue. determined by Q-RT-PCR analysis of human alpha satellite and avian GAPDH DNA normalized per cell. Very similar results were obtained with Revlimid. The results presented suggest that with further work the turkey embryo model may provide an affordable, rapid and practical xenograft system in vivo for studying the biology of MM, for affordably testing MM therapies, as well for developing a new method for individualized patient screening for response or resistance to particular therapeutic agents. 1. Taizi M, Deutsch VR, Leitner A, Ohana A, Goldstein RS. A novel and rapid in vivo system for testing therapeutics on human leukemias. Exp Hematol. 2006;34:1698-1708. 2. Grinberg I, Reis A, Ohana A, et al. Engraftment of human blood malignancies to the turkey embryo: a robust new in vivo model. Leuk Res. 2009;33:1417-1426. Disclosures: No relevant conflicts of interest to declare.


2006 ◽  
Vol 26 (7) ◽  
pp. 2615-2625 ◽  
Author(s):  
John E. Pimanda ◽  
Lev Silberstein ◽  
Massimo Dominici ◽  
Benjamin Dekel ◽  
Mark Bowen ◽  
...  

ABSTRACT Blood and vascular cells are generated during early embryogenesis from a common precursor, the hemangioblast. The stem cell leukemia gene (SCL/tal 1) encodes a basic helix-loop-helix transcription factor that is essential for the normal development of blood progenitors and blood vessels. We have previously characterized a panel of SCL enhancers including the +19 element, which directs expression to hematopoietic stem cells and endothelium. Here we demonstrate that SCL is expressed in bone primordia during embryonic development and in adult osteoblasts. Despite consistent expression in cells of the osteogenic lineage, SCL protein is not required for bone specification of embryonic stem cells. In transgenic mice, the SCL +19 core enhancer directed reporter gene expression to vascular smooth muscle and bone in addition to blood and endothelium. A 644-bp fragment containing the SCL +19 core enhancer was active in both blood and bone cell lines and was bound in vivo by a common array of Ets and GATA transcription factors. Taken together with the recent observation that a common progenitor can give rise to blood and bone cells, our results suggest that the SCL +19 enhancer targets a mesodermal progenitor capable of generating hematopoietic, vascular, and osteoblastic progeny.


2005 ◽  
Vol 79 (15) ◽  
pp. 10088-10092 ◽  
Author(s):  
Nanae Harashima ◽  
Ryuji Tanosaki ◽  
Yukiko Shimizu ◽  
Kiyoshi Kurihara ◽  
Takao Masuda ◽  
...  

ABSTRACT We previously reported that Tax-specific CD8+ cytotoxic T lymphocytes (CTLs), directed to single epitopes restricted by HLA-A2 or A24, expanded in vitro and in vivo in peripheral blood mononuclear cells (PBMC) from some adult T-cell leukemia (ATL) patients after but not before allogeneic hematopoietic stem cell transplantation (HSCT). Here, we demonstrated similar Tax-specific CTL expansion in PBMC from another post-HSCT ATL patient without HLA-A2 or A24, whose CTLs equally recognized two newly identified epitopes, Tax88-96 and Tax272-280, restricted by HLA-A11, suggesting that these immunodominant Tax epitopes are present in the ATL patient in vivo.


Genomics ◽  
2019 ◽  
Vol 111 (6) ◽  
pp. 1566-1573 ◽  
Author(s):  
Jeane Silva ◽  
Chang-Sheng Chang ◽  
Tianxiang Hu ◽  
Haiyan Qin ◽  
Eiko Kitamura ◽  
...  

2006 ◽  
Vol 18 (2) ◽  
pp. 208 ◽  
Author(s):  
A. S. Lima ◽  
S. A. Malusky ◽  
M. R. B. Mello ◽  
S. J. Lane ◽  
J. R. Rivera ◽  
...  

A primary concern in stem cell biology is that observations made in vitro may be an artifact of the in vitro culture environment. In vitro derived stem cells can be implanted into the environment from which they are derived so that their response to physiological conditions may be observed. Several important cellular characteristics need to be examined following the cell's reintroduction to the in vivo environment, including the potential for differentiation, proliferative ability, and life span. Studying implanted stem cells will assist in determining the potential for stem cell use in clinical therapies and provide further understanding of the role adult stem cells have in the adult body. Currently, the scientific literature is lacking a detailed description of the cellular response of adipose-derived stem cells (ADSCs) reintroduced to their exact tissue of origin. Thus, the aim of this study was to evaluate porcine ADSC growth in vivo and to analyze cell differentiation in vivo following injection of undifferentiated ADSCs into subcutaneous fat. Subcutaneous adipose tissue was isolated from the back fat of male pigs (11 months of age) and digested with 0.075% collagenase at 37�C for 90 min. The digested tissue was centrifuged at 200g for 10 min to obtain a cell pellet. The pellet was re-suspended with DMEM and the ADSCs were plated onto 75 cm2 flasks (5000-10 000 cells per cm2) and cultured in DMEM supplemented with 10% fetal bovine serum (FBS) and 1% gentamicin. Passage 3 ADSCs were labeled with fluorescent dye (PKH26; Sigma, St. Louis, MO, USA) and sorted by flow cytometry. After sorting, positive cells were washed and re-suspended in culture medium. For transplantation, 100 �L of cell suspension in DMEM containing one of four cell concentrations (0 (control); 30 000; 300 000; and 900 000 cells) were placed in a 1-mL syringe and injected into the subcutaneous back fat of recipient pigs (n = 2). Each pig had previously been tattooed with 12 13 � 13 squares to mark injection sites. The treatments were replicated three times within each animal. Two and three weeks after transplantation, animals were euthanized, the back fat containing the transplantation site was harvested, and the cells were disaggregated as described above. The buoyant adipocytes and pelleted ADSCs cells were then analyzed by flow cytometry. The results indicated that there were dose- and time-dependent increases in labeled ADSCs and labeled adipocytes in the fat samples with increasing cell number (from 0 to 300 000 cells). There was, however, a decrease in labeled ADSCs at the 900 000-cell dose, which is likely due to excess cells being transplanted or an immune reaction. Both of these aspects are currently being evaluated. In conclusion, undifferentiated ADSCs from swine can be isolated from and returned to the subcutaneous adipose layer and differentiate into mature adipocytes. This work was supported by the Council for Food and Agricultural Research (C-FAR) Sentinel Program, University of Illinois.


2019 ◽  
Vol 40 (Supplement_1) ◽  
Author(s):  
D Mori ◽  
S Miyagawa ◽  
T Kawamura ◽  
H Hata ◽  
T Ueno ◽  
...  

Abstract Background Although transplantation of human Adipose-derived Mesenchymal stem cell (hADSC) shows efficacy in the treatment of ischemic cardiomyopathy, its therapeutic mechanisms have not been fully elucidated. It has been already reported that mitochondria transfer to recipient cells have impact on resistance to injury and tissue regeneration, however this phenomenon has not been elucidated in the damaged heart. Therefore, we hypothesized that ADSC transfer own mitochondria to cardiomyocytes in-vivo and in-vitro under ischemic condition, resulting in the functional recovery of cardiomyocyte. Method and result Transplantation of hADSC (group A) to the heart surface or sham operation (group C) was performed in rats that were subjected to LAD ligation 2 weeks prior to the treatment (n=10 each). The number of transplant cell was 1x106/body. Three days after transplantation, transferred hADSCs' mitochondria were observed in recipient cardiomyocytes histologically (Figure). Quantitative PCR analysis revealed that mitochondrial genome of recipient myocytes increased over time. The cardiac function assessed with echocardiography was significantly better in group A. Furthermore, live-imaging of hADSC transplantation revealed the suspected transfer of mitochondria to beating heart. In-vitro, the co-culture of rat cardiomyocytes (rCM) and hADSC was observed with time-lapse photography and demonstrated mitochondrial transfer under the hypoxic condition. The measuring the oxygen consumption rate (OCR) of these cells showed that OCR of rCM was reinforced by co-culture with hADSC conspicuously. Figure 1 Conclusion Mitochondrial transfer from hADSC to rCM was suggested in-vivo and in-vitro ischemic condition and suspected to be related to functional recovery of ischemic cardiomyocyte.


Sign in / Sign up

Export Citation Format

Share Document