scholarly journals Characterization of Monoclonal Antibodies That Recognize Canine CD34

Blood ◽  
1998 ◽  
Vol 91 (6) ◽  
pp. 1977-1986 ◽  
Author(s):  
Peter A. McSweeney ◽  
Katherine A. Rouleau ◽  
Philip M. Wallace ◽  
Benedetto Bruno ◽  
Robert G. Andrews ◽  
...  

Abstract Using a polyclonal antiserum against canine CD34, we previously found that CD34 is expressed on canine bone marrow progenitor cells in a manner analogous to that found in humans. To further characterize CD34+ cells and to facilitate preclinical canine stem cell transplant studies, monoclonal antibodies (MoAbs) were raised to CD34. A panel of 10 MoAbs was generated that reacted with recombinant CD34 and with CD34+ cell lines and failed to react with CD34− cell lines. Binding properties of five purified MoAbs were determined by BIAcore analysis and flow cytometric staining, and several MoAbs showed high affinity for CD34. Two antibodies, 1H6 and 2E9, were further characterized, and in flow cytometry studies typically 1% to 3% of stained bone marrow cells were CD34+. Purified CD34+ bone marrow cells were 1.8- to 55-fold enriched for colony-forming unit–granulocyte-macrophage and for long-term culture initiating cells as compared with bone marrow mononuclear cells, whereas CD34− cells were depleted of progenitors. Three autologous transplants were performed with CD34+ cell fractions enriched by immunomagnetic separation. After marrow ablative total body irradiation (920 cGy), prompt hematopoietic recovery was seen with transplanted cell doses of ≤1.1 × 107 /kg that were 29% to 70% CD34+. Engraftment kinetics were similar to those of dogs previously transplanted with approximately 10- to 100-fold more unmodified autologous marrow cells. This suggests that CD34+ is a marker not only of canine bone marrow progenitors but also for cells with radioprotective or marrow repopulating function in vivo. MoAbs to CD34 will be valuable for future studies of canine hematopoiesis and preclinical studies concerning stem cell transplantation, gene therapy, and ex vivo progenitor cell expansion.

Blood ◽  
1998 ◽  
Vol 91 (6) ◽  
pp. 1977-1986 ◽  
Author(s):  
Peter A. McSweeney ◽  
Katherine A. Rouleau ◽  
Philip M. Wallace ◽  
Benedetto Bruno ◽  
Robert G. Andrews ◽  
...  

Using a polyclonal antiserum against canine CD34, we previously found that CD34 is expressed on canine bone marrow progenitor cells in a manner analogous to that found in humans. To further characterize CD34+ cells and to facilitate preclinical canine stem cell transplant studies, monoclonal antibodies (MoAbs) were raised to CD34. A panel of 10 MoAbs was generated that reacted with recombinant CD34 and with CD34+ cell lines and failed to react with CD34− cell lines. Binding properties of five purified MoAbs were determined by BIAcore analysis and flow cytometric staining, and several MoAbs showed high affinity for CD34. Two antibodies, 1H6 and 2E9, were further characterized, and in flow cytometry studies typically 1% to 3% of stained bone marrow cells were CD34+. Purified CD34+ bone marrow cells were 1.8- to 55-fold enriched for colony-forming unit–granulocyte-macrophage and for long-term culture initiating cells as compared with bone marrow mononuclear cells, whereas CD34− cells were depleted of progenitors. Three autologous transplants were performed with CD34+ cell fractions enriched by immunomagnetic separation. After marrow ablative total body irradiation (920 cGy), prompt hematopoietic recovery was seen with transplanted cell doses of ≤1.1 × 107 /kg that were 29% to 70% CD34+. Engraftment kinetics were similar to those of dogs previously transplanted with approximately 10- to 100-fold more unmodified autologous marrow cells. This suggests that CD34+ is a marker not only of canine bone marrow progenitors but also for cells with radioprotective or marrow repopulating function in vivo. MoAbs to CD34 will be valuable for future studies of canine hematopoiesis and preclinical studies concerning stem cell transplantation, gene therapy, and ex vivo progenitor cell expansion.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4836-4836
Author(s):  
Xiaofei Liu ◽  
Dan Yang ◽  
Shumin Fang ◽  
Yang Geng ◽  
Qianhao Zhao ◽  
...  

Abstract Traditional mouse models for hematopoietic stem cell transplant (HSCT) require whole body irradiation to ablate the hematopoietic cells in recipients, with the defect of disturbing non-hematopoietic cells and introducing potential tumorgenesis in a nonautonomous manner. Here, we use a novel approach to produce mice that whole hematopoietic-specific ablation can be conditionally achieved without global body irradiation. Briefly, a Diphtheria toxin receptor (DTR)-GFP reporter element was targeted into the ROSA26 locus to produce DTR-GFP reporter mice, with a loxp-stop-loxp cassette. Then the DTR-GFP reporter mice were crossed to Vav-Cre mice to produce double transgenic mice (DTR-GFP mice). We injected DT to ablate the bone marrow cells from DTR-GFP mice, and transplant WT bone marrow cells into them. Our data showed that all hematopoietic cells including hematopoietic stem cells, Myeloid, lymphoid lineages are GFP positive in DTR-GFP mice. A single dose of DT can kill all the hematopoietic cells from DTR-GFP mice. One month later, WT bone marrow cells were successfully engrafted into the DTR-GFP recipients without irradiation. We are using this model to re-evaluate some leukemia models that irradiated bone marrow niches might be involved in the tumorigenesis.Thus, we establish a de novel HSCT approach without irradiated myeloablation, which will benefit studies of hematopoiesis, leukemogenesis, hematopoietic stem cell niche, as well as other types of tissue transplants that need ablation of recipient hematopoietic system or immune system. Disclosures: Peng: Biocytogen: Employment, Membership on an entity’s Board of Directors or advisory committees. Shen:Biocytogen: Employment, Membership on an entity’s Board of Directors or advisory committees.


Blood ◽  
2012 ◽  
Vol 120 (9) ◽  
pp. 1933-1941 ◽  
Author(s):  
Satoru Otsuru ◽  
Patricia L. Gordon ◽  
Kengo Shimono ◽  
Reena Jethva ◽  
Roberta Marino ◽  
...  

Abstract Transplantation of whole bone marrow (BMT) as well as ex vivo–expanded mesenchymal stromal cells (MSCs) leads to striking clinical benefits in children with osteogenesis imperfecta (OI); however, the underlying mechanism of these cell therapies has not been elucidated. Here, we show that non–(plastic)–adherent bone marrow cells (NABMCs) are more potent osteoprogenitors than MSCs in mice. Translating these findings to the clinic, a T cell–depleted marrow mononuclear cell boost (> 99.99% NABMC) given to children with OI who had previously undergone BMT resulted in marked growth acceleration in a subset of patients, unambiguously indicating the therapeutic potential of bone marrow cells for these patients. Then, in a murine model of OI, we demonstrated that as the donor NABMCs differentiate to osteoblasts, they contribute normal collagen to the bone matrix. In contrast, MSCs do not substantially engraft in bone, but secrete a soluble mediator that indirectly stimulates growth, data which provide the underlying mechanism of our prior clinical trial of MSC therapy for children with OI. Collectively, our data indicate that both NABMCs and MSCs constitute effective cell therapy for OI, but exert their clinical impact by different, complementary mechanisms. The study is registered at www.clinicaltrials.gov as NCT00187018.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3276-3276 ◽  
Author(s):  
Anupriya Agarwal ◽  
Ryan Mackenzie ◽  
Dorian LaTocha ◽  
Kavin Vasudevan ◽  
Eduardo Firpo ◽  
...  

Abstract Abstract 3276 Poster Board III-1 Background: Several studies have indicated that BCR-ABL causes cell cycle defects by interfering with the cell cycle regulatory functions of p27, a Cyclin dependent kinase (Cdk) inhibitor and tumor suppressor. Studies in BCR-ABL positive cell lines have shown that BCR-ABL promotes proteasomal degradation of p27 in a pathway that involves the SCFSKP2 ubiquitin ligase, while cytoplasmic mislocalization has been described in primary CML cells. It has been suggested that the principal effect of this cytoplasmic mislocalization is to remove p27 from the nucleus, thereby relieving Cdks from p27 inhibition. However, recent studies have shown that a p27 mutant (p27CK-), that cannot bind to Cdks or Cyclins, actively contributes to oncogenesis. This raises the question as to whether cytoplasmic mislocalization of p27 in CML cells may in fact promote leukemogenesis rather than merely compromise Cdk inhibition. We therefore hypothesized that the net contribution of p27 in CML is to promote leukemogenesis due to the oncogenic activity of cytoplasmic p27. Experimental approach and results: We determined p27 localization in BCR-ABL positive cell lines and CD34+ progenitor cells from newly diagnosed chronic phase CML patients (N=7) and from CML patients in blast crisis (N=2) by immunoblotting of nuclear and cytoplasmic cellular fractions. We found that p27 is predominantly cytoplasmic in most CML cell lines and in CD34+ cells from 8/9 (89%) patient samples, including patients in blastic phase. Cytoplasmic localization of p27 in CD34+ cells from CML patients was also confirmed by immunofluorescence analysis. Further, we observed that inhibition of BCR-ABL kinase by imatinib, an Abl kinase inhibitor increased nuclear p27 in all cell lines tested and in 4/9 patient samples (3/7 chronic phase and 1/2 blastic phase samples). However, we did not observe a substantial change in the cytoplasmic p27 levels. Similar results were obtained in Ba/F3 and 32D murine hematopoietic cell lines expressing BCR-ABL when compared with the respective parental cells. Further, SKP2 was up-regulated in CD34+ cell from CML patients as compared to the normal patients consistent withSKP2 mediated down-regulation of nuclear p27. These data suggest that nuclear but not cytoplasmic p27 levels are predominantly regulated by BCR-ABL kinase activity. To test whether p27 is crucial for BCR-ABL-driven leukemia, we compared leukemogenesis between recipients of BCR-ABL transduced p27+/+ and p27-/- bone marrow. Mice transplanted with BCR-ABL infected p27-/- marrow had significantly longer median survival (70 days, range 48-150 days) compared to recipients of p27+/+ marrow (37 days, range 14-56 days) (p=0.0123). To exclude that this difference was related to the differences in homing and engraftment capabilities of p27+/+ and p27-/- bone marrow cells, we compared short term homing and long term engraftment of p27+/+ and p27-/- bone marrow cells transplanted into wild-type recipients and found no differences. These data suggest that the net contribution of p27 to BCR-ABL-mediated leukemogenesis is positive. Further, to investigate the contribution of nuclear p27 to leukemogenesis, we utilized marrow from p27S10A mice in the murine CML model. In p27S10A mice, p27 is nuclear to to abrogation of the phosphorylation site implicated in nuclear export. We injected BCR-ABL transduced bone marrow cells of p27S10A and p27+/+ mice into wild-type recipients and compared the disease progression. We observed that mice transplanted with BCR-ABL infected p27S10A marrow had significantly longer median survival (28 days, range 23-79 days) compared to the recipients of p27+/+ marrow (23 days, range 21-38 days) (p=0.0139). This data is consistent with nuclear tumor suppressor function of p27. Combined with the data above, this suggests that cytoplasmic p27 promotes BCR-ABL mediated leukemogenesis. Conclusions: Our data suggest that though nuclear p27 functions as a tumor suppressor, the net contribution of p27 in CML might be oncogenic due to an oncogenic role of the increased cytoplasmic p27. Restoring nuclear p27 or reducing cytoplasmic p27 may be therapeutically useful in malignancies with low nuclear and high cytoplasmic p27 expression. Disclosures: Druker: OHSU patent #843 - Mutate ABL Kinase Domains: Patents & Royalties; MolecularMD: Equity Ownership; Roche: Consultancy; Cylene Pharmaceuticals: Consultancy; Calistoga Pharmaceuticals: Consultancy; Avalon Pharmaceuticals: Consultancy; Ambit Biosciences: Consultancy; Millipore via Dana-Farber Cancer Institute: Patents & Royalties; Novartis, ARIAD, Bristol-Myers Squibb: Research Funding. Deininger:Genzyme: Research Funding; BMS: Consultancy; Novartis: Consultancy, Honoraria; Ariad : Research Funding.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1893-1893
Author(s):  
Carolien M Woolthuis ◽  
Annet Z Vos ◽  
Gerwin Huls ◽  
Joost TM de Wolf ◽  
Jan J. Schuringa ◽  
...  

Abstract Abstract 1893 Autologous stem cell transplantation (ASCT) allows the application of high-dose chemotherapy and is included in the standard treatment regimens for multiple myeloma and relapsing lymphoma. The application of ASCT has considerably improved treatment outcome but in 30–50% of the patients the underlying malignant disorder relapses. In that case treatment options are limited, in part due to a diminished capacity of the transplanted bone marrow to tolerate chemotherapy. The transplanted bone marrow seems to be more vulnerable to chemotoxic stress. This is supported by our recent observations that progenitor cells from post-ASCT bone marrow demonstrate altered phenotypic and functional properties (Woolthuis et al. BMT 2011:110-115). In the present study, a more detailed examination of the hematopoietic stem cell (HSC) compartment post-ASCT was performed. The examined bone marrow cells were obtained from patients with normal peripheral blood cell counts one year after ASCT. First it was studied whether stem cell quiescence had been changed due to the ASCT procedure. Therefore the percentage of cells in the G0 phase was measured by staining the cells with Hoechst and Pyronin Y followed by flowcytometric analysis. Both the stem cell (CD34+/CD38low) and progenitor (CD34+/CD38+) fractions were analyzed. Post-ASCT bone marrow cells (n=6) were compared with normal bone marrow cells (n=9) and mobilized peripheral blood stem cells (PBSC) (n=7). Interestingly, post-ASCT bone marrow contained a significantly lower percentage of quiescent cells in the CD34+/CD38low fraction compared to normal bone marrow (mean percentage 23.6% (95%CI: 6.5–40.8) vs. 48.6% (95%CI: 31.4–65.9), p=0.045). In contrast no differences were observed in the CD34+/CD38+ fraction. To examine whether the loss of stem cell quiescence is associated with loss of stem cell function, stem cell frequency was analyzed in vitro by the long-term colony-initiating cell assay. These analyses revealed a strongly decreased stem cell frequency in post-ASCT CD34+ cells compared to normal CD34+ bone marrow cells (mean frequency 0.0016 (95%CI: 0.0003–0.0028) vs. 0.0206 (95%CI: 0.0162–0.0250), p=0.002). Importantly, CD34+ PBSC cells demonstrated a stem cell frequency comparable with the frequency observed in normal bone marrow, suggesting that the decrease in stem cell frequency observed in post-ASCT bone marrow can not simply be explained by the previous chemotherapy and the mobilization procedure. To obtain more insight into the mechanisms explaining our observations, levels of reactive oxygen species (ROS) were measured by flowcytometry. Comparable ROS levels were observed in post-ASCT and normal bone marrow CD34+/CD38low cells while the CD34+/CD38+ post-ASCT cells demonstrated significantly higher ROS levels compared to normal bone marrow (p=0.01). In addition significant higher ROS levels were observed in CD34+/CD38low PBSC compared to normal bone marrow (p=0.043). To define pathways associated with altered quiescence and/or ROS production, gene expression analysis was performed comparing CD34+ cells from post-ASCT bone marrow (n=6), normal bone marrow (n=31) and PBSC (n=5). Preliminary analysis reveals an upregulation in post-ASCT CD34+ cells of genes involved in cell cycle compared to normal bone marrow. In contrast, cell cycle genes appeared to be downregulated in CD34+ PBSC cells. In conclusion, our data indicate that the diminished regenerative capacity of bone marrow post-ASCT is likely related to a loss of stem cell quiescence and enhanced ROS production by progenitor cells which will make the bone marrow cells more vulnerable for the effects of cytotoxic stress. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1998 ◽  
Vol 92 (7) ◽  
pp. 2269-2279 ◽  
Author(s):  
Kevin D. Bunting ◽  
Jacques Galipeau ◽  
David Topham ◽  
Ely Benaim ◽  
Brian P. Sorrentino

Attempts to expand repopulating hematopoietic cells ex vivo have yielded only modest amplification in stem cell numbers. We now report that expression of an exogenous human multi-drug resistance 1 (MDR1) gene enables dramatic ex vivo stem cell expansion in the presence of early acting hematopoietic cytokines. Bone marrow cells were transduced with retroviral vectors expressing either the MDR1 gene or a variant of human dihydrofolate reductase (DHFR), and then expanded for 12 days in the presence of interleukin-3 (IL-3), IL-6, and stem cell factor. When these cells were injected into nonirradiated mice, high levels of long-term engraftment were only seen with MDR1-transduced grafts. To verify that expansion of MDR1-transduced repopulating cells had occurred, competitive repopulation assays were performed using MDR1 expanded grafts. These experiments showed progressive expansion of MDR1-transduced repopulating cells over the expansion period, with a 13-fold overall increase in stem cells after 12 days. In all of the experiments, mice transplanted with expanded MDR1-transduced stem cells developed a myeloproliferative disorder characterized by high peripheral white blood cell counts and splenomegaly. These results show that MDR1-transduced stem cells can be expanded in vitro using hematopoietic cytokines without any drug selection, but enforced stem cell self-renewal divisions can have adverse consequences.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1631-1631
Author(s):  
Andrew G. Aprikyan ◽  
Oscar Penate ◽  
Vahagn Makaryan

Abstract Myelokathexis (MK) is a rare congenital disorder characterized by hypercellular marrow and severe chronic neutropenia. In some, but not all patients there is an association of Warts, Hypogammaglobulinemia, and Infections with Myelokathexis (WHIM syndrome). We and others reported that bone marrow myeloid cells in MK exhibit characteristic apoptotic features such as condensed chromatin, cytoplasmic blebbing, and cellular fragmentation. FACS analysis also demonstrated an impaired survival of myeloid cells in MK that was associated with reduced level of Bcl-X expression. Several heterozygous mutations in the CXCR4 gene have been identified in most of the families with autosomal dominant WHIM syndrome. CXCR4 is a G-protein coupled chemokine receptor and its interaction with SDF-1 ligand plays an important role in homing and mobilization of hematopoietic cells. Recently, it has been demonstrated that expression of mutant CXCR4 in human CD34+ cells results in reduced receptor internalization, enhanced calcium flux and enhanced migration of transduced CD34+ cells. However, the mechanism of mutant CXCR4-mediated neutropenia in MK remains largely unknown. We examined 6 patients representing 4 unrelated families with MK and found that all affected family members harbor heterozygous mutations in the CXCR4 gene including a previously reported R334ter mutation. We identified two novel mutations that result in frame shifts and a premature stop codon in the cytoplasmic domain of CXCR4. Expression of CXCR4 mutants in human promyelocytic HL-60 cells resulted in massive apoptosis similar to that reported for bone marrow-derived myeloid cells from MK patients. Specifically, more than 50% of HL-60 cells transfected with previously reported R334ter and novel truncation mutants were positive for annexin V, whereas 20±6% of apoptotic annexin V-positive cells were observed in control cells transfected with normal CXCR4. Impaired cell survival appeared to be due to aberrant dissipation of mitochondrial membrane potential as flow cytometry analysis revealed significantly increased DIOC6-staining in cells expressing CXCR4 mutants compared with control cells expressing normal CXCR4 (p<0.02). Moreover, the expression of CXCR4 mutants but not of the wild type form led to a significant increase in directional motility of myeloid cells towards SDF-1 (p<0.01), similar to the enhanced chemotaxis of blood mononuclear cells observed in our MK patients. These data indicate that our cellular model closely recapitulates the myelokathexis phenotype. Premature apoptosis, but not enhanced chemotaxis triggered by mutant CXCR4 was reduced to near-normal level by caspase-specific inhibitor zVAD-fmk. Interestingly, the mutant CXCR4-induced increase in directional motility to SDF-1, but not accelerated apoptosis was normalized upon treatment with protein kinase Cξ-specific inhibitor. These data suggest that accelerated apoptosis and increased chemotaxis are two independent pathways activated by mutant CXCR4. Importantly, treatment of primary blood mononuclear cells from 3 MK patients with PKCξ inhibitor restored the abnormal chemotactic properties to the levels comparable to that of treated control cells from healthy volunteers. Further studies needed for assessing the therapeutic potential of PKCξ-specific inhibitor for normalizing the impaired mobilization of bone marrow cells in MK. Our data also suggest that the PKCξ-specific inhibitor may be effective for mobilization of human hematopoietic stem cells.


Blood ◽  
1998 ◽  
Vol 92 (7) ◽  
pp. 2269-2279 ◽  
Author(s):  
Kevin D. Bunting ◽  
Jacques Galipeau ◽  
David Topham ◽  
Ely Benaim ◽  
Brian P. Sorrentino

Abstract Attempts to expand repopulating hematopoietic cells ex vivo have yielded only modest amplification in stem cell numbers. We now report that expression of an exogenous human multi-drug resistance 1 (MDR1) gene enables dramatic ex vivo stem cell expansion in the presence of early acting hematopoietic cytokines. Bone marrow cells were transduced with retroviral vectors expressing either the MDR1 gene or a variant of human dihydrofolate reductase (DHFR), and then expanded for 12 days in the presence of interleukin-3 (IL-3), IL-6, and stem cell factor. When these cells were injected into nonirradiated mice, high levels of long-term engraftment were only seen with MDR1-transduced grafts. To verify that expansion of MDR1-transduced repopulating cells had occurred, competitive repopulation assays were performed using MDR1 expanded grafts. These experiments showed progressive expansion of MDR1-transduced repopulating cells over the expansion period, with a 13-fold overall increase in stem cells after 12 days. In all of the experiments, mice transplanted with expanded MDR1-transduced stem cells developed a myeloproliferative disorder characterized by high peripheral white blood cell counts and splenomegaly. These results show that MDR1-transduced stem cells can be expanded in vitro using hematopoietic cytokines without any drug selection, but enforced stem cell self-renewal divisions can have adverse consequences.


Blood ◽  
2010 ◽  
Vol 116 (15) ◽  
pp. 2676-2683 ◽  
Author(s):  
Alevtina D. Domashenko ◽  
Gwenn Danet-Desnoyers ◽  
Alissa Aron ◽  
Martin P. Carroll ◽  
Stephen G. Emerson

Abstract Retroviral overexpression of NF-Ya, the regulatory subunit of the transcription factor NF-Y, activates the transcription of multiple genes implicated in hematopoietic stem cell (HSC) self-renewal and differentiation and directs HSCs toward self-renewal. We asked whether TAT-NF-Ya fusion protein could be used to transduce human CD34+ cells as a safer, more regulated alternative approach to gene therapy. Here we show that externally added recombinant protein was able to enter the cell nucleus and activate HOXB4, a target gene of NF-Ya, using real-time polymerase chain reaction RNA and luciferase-based protein assays. After TAT-NF-Ya transduction, the proliferation of human CD34+ cells in the presence of myeloid cytokines was increased 4-fold. Moreover, TAT-NF-Ya-treated human primary bone marrow cells showed a 4-fold increase in the percentage of huCD45+ cells recovered from the bone marrow of sublethally irradiated, transplanted NOD-Scid IL2Rγnull mice. These data demonstrate that TAT-peptide therapies are an alternative approach to retroviral stem cell therapies and suggest that NF-Ya peptide delivery should be further evaluated as a tool for HSC/progenitors ex vivo expansion and therapy.


Sign in / Sign up

Export Citation Format

Share Document