Oncostatin M Enhances Hematopoietic Stem Cell Homing and Engraftment

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 31-31
Author(s):  
Hannah K Rasmussen ◽  
Frankie Wong ◽  
Tiffany Tate ◽  
David T. Scadden ◽  
Jonathan Hoggatt

Abstract Hematopoietic stem cell (HSC) transplant is widely used to treat hematologic, metabolic, and malignant diseases. HSCs reside within a microenvironmental stem cell niche; the cellular makeup of which is complex, with contributions from endothelial, mesenchymal, and mature hematopoietic cells. Despite the longstanding use of HSC transplantation, there are remarkably few, if any, therapeutics that are available today that can be administered to the recipient of a transplant to enhance hematopoietic engraftment. We believe that revealing the molecular toolkit that naturally exists within the niche provides a rich source of potential novel therapies. Recently, three independent groups (Winkler et al., 2010; Christopher et al., 2011; Chow et al., 2011) have implicated macrophages as a key regulator in G-CSF-mediated mobilization of HSCs. These groups demonstrated that macrophages express the G-CSF receptor, and that depletion of macrophages leads to niche attenuation, reduced levels of stromal derived factor-1 (SDF-1), and mobilization of HSCs. This data implied that macrophages produce a positive supporting factor(s) in the niche to support HSC retention, though none of these prior reports were able to identify the key molecule(s). We now report that Oncostatin M (OSM) is a macrophage-produced protein that regulates HSC retention within the niche. Initial identification was achieved utilizing an in vitro co-culture screening system with macrophage-conditioned media and response of MS-5 stromal cells as measured by SDF-1 production. As previously demonstrated, macrophages enhanced SDF-1 production; however, when OSM production was reduced via shRNA knockdown, or the OSM receptor was knocked down in MS-5 cells or blocked with antibody, the enhanced SDF-1 production was abrogated. Intriguingly, this effect was specific to OSM, as changes in IL-6 or LIF signaling, both members of a similar gp130 signaling family along with OSM, did not have any effects. When mice were treated G-CSF, significant reductions in bone marrow SDF-1 levels and mobilization resulted. However, when mice were co-treated with OSM, levels of bone marrow SDF-1 remained the same as untreated mice (n=10 mice per group, 2 independent expts, P<0.01). Similarly, when macrophages were depleted in vivo via clodronate-loaded liposome treatment, significant HSC mobilization occurred, which was blocked with co-treatment of OSM (n=10 mice per group, 2 independent expts, P<0.01). These results now elucidate a previously unknown key regulatory mechanism governing G-CSF and macrophage mediated mobilization of HSCs. We hypothesized that this new biologic insight could be leveraged as a novel therapeutic strategy to enhance HSC homing and engraftment. To test this, we conducted a series of experiments in which we conditioned mice with myeloablative irradiation and treated with either vehicle control or recombinant OSM (0.5ug per injection) every 6 hours for 48 hours at which point mice received a transplant of bone marrow cells and were subsequently analyzed post-transplant. Mice pre-treated with OSM prior to transplantation exhibited a 2-fold increase in HSC homing compared to vehicle control groups (n=10 mice per group, 3 independent expts, P<0.05). Furthermore, these homed progenitors demonstrated remarkably enhanced hematopoietic expansion in OSM treated mice, as demonstrated by increased numbers of colony forming units (CFUs) from bone marrow assessed days 4, 7, 11, and 14 post-transplant (n=10 mice per group, P<0.01). Excitingly, using a limited cell number transplant, mimicking settings of single cord blood unit transplantation in adults or other settings of limited HSC number, we demonstrate enhanced hematopoietic engraftment and survival in OSM pre-treated mice, with 100 percent survival compared to 50 percent survival of mice treated with vehicle control (n=20 per group, P<0.001). Collectively, our results demonstrate how illumination of endogenous regulatory mechanisms within the hematopoietic niche can reveal molecular agents and pathways with potential to serve as new therapeutic agents in the clinic. Specifically, we identify OSM as the key regulatory molecule governing the G-CSF-macrophage mediated mobilization of HSCs, and describe for the first time a novel therapeutic approach using recombinant OSM as a therapeutic to meet a currently unmet clinical need for HSC transplantation. Disclosures Hoggatt: Harvard University: Patents & Royalties.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4321-4321
Author(s):  
Miguel Ganuza Fernandez ◽  
Per Holmfeldt ◽  
Himangi Marathe ◽  
Trent Hall ◽  
Jennifer Pardieck ◽  
...  

Abstract Introduction: Hematopoiesis involves the hierarchical generation of the major blood lineages from a common ancestor, the Hematopoietic Stem Cell (HSC). HSC also have the intrinsic ability to repopulate an ablated hematopoietic compartment when introduced into the periphery of a recipient. This has allowed Hematopoietic Stem Cell transplantation (HSCT) to be used as a cell therapy over the last 45 years, benefiting thousands of patients. Unfortunately many patients succumb to disease while waiting for an adequate donor. Others have to undergo unrelated donor transplants, putting themselves at a risk of developing graft-versus-host disease. Improving HSC engraftment could ameliorate transplant morbidity. Thus, understanding mechanisms regulating HSC engraftment is key. Results: We used our recently published gene expression profiles of developing HSC and other public databases to prioritize 58 genes as putative regulators of adult HSC function. We confirmed by qRT-PCR that 51/58 candidates were enriched for gene expression in Lineage-Sca-1+c-Kit+ (LSK) bone marrow cells relative to downstream progeny, suggesting a role in hematopoietic stem and progenitor cells (HSPC). To functionally assay a role for each gene of interest (GOI) in HSC engraftment, we designed and validated ≥2 independent shRNAs/GOI that effected a >75% knockdown in gene expression in LSK cells. LSK cells were lentivirally transduced with control or individual gene-specific shRNAs and transplanted into lethally irradiated recipients along with mock-transduced LSK competitor cells congenic at the CD45 allele. In contrast to previous functional screens, transplant was performed within 24-hours of LSK cell isolation, avoiding extensive ex vivo culture. This minimal manipulation allowed us to detect genes critical for efficient HSC engraftment. Peripheral blood chimerism was analyzed for at least 16 weeks post-transplant. The major bone marrow hematopoietic compartments were also analyzed. 17 of 48 genes tested were identified as necessary for optimal HSPC engraftment (i.e. knockdown induced a significant loss of repopulation) and the knockdown of three genes enhanced HSPC repopulation. shRNAs targeting each “Hit” were interrogated ex vivo for non-specific effects on LSK cell viability and expansion. A 2° screen was performed to validate the results of this primary screen. Here, CD45.2 LSK cells transduced with control or individual gene-specific shRNAs were sorted 48 hours post-transduction for mCherry+ cells and then transplanted into lethally irradiated mice with mock-transduced and mock-sorted CD45.1 congenic LSK cells. 18 “Hits” were confirmed to perturb HSC repopulating potential in this 2° screen, including three whose loss enhanced HSPC repopulation. The transcription factor, Foxa3, is one hit identified here as necessary for HSC repopulation. We further found that that Foxa3-/- bone marrow displays a significant loss of repopulating potential >16 weeks post-transplant, confirming the results of our screen. As Foxa3-/- long-term HSC also display reduced colony forming potential in vitro and fail to contribute to downstream progenitor compartments in transplant recipients, we propose that Foxa3 is a novel regulator of HSC differentiation post-transplant. Foxa3 has never before been implicated in hematopoiesis or HSPC biology. Conclusions: Our novel functional screen has revealed 15 genes required for optimal HSPC engraftment and three genes whose knockdown improved HSPC engraftment. We further validated Foxa3 as a novel regulator of HSC activity by demonstrating that Foxa3-/- HSC are also deficient in repopulating activity. We are currently investigating the molecular mechanism of Foxa3’s role in HSC and, given that Foxa genes are known transcriptional pioneering factors, pursuing the hypothesis that Foxa3 functions as a novel epigenetic regulator of HSC activation and differentiation. Each gene identified in our screen represents a window into the discovery of novel mechanisms regulating HSC biology and engraftment. Disclosures No relevant conflicts of interest to declare.


Leukemia ◽  
2021 ◽  
Author(s):  
Kavita Bisht ◽  
Crystal McGirr ◽  
Seo-Youn Lee ◽  
Hsu-Wen Tseng ◽  
Whitney Fleming ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 361-361 ◽  
Author(s):  
Laura A. Paganessi ◽  
Stephanie A. Gregory ◽  
Henry C. Fung ◽  
Kent W. Christopherson

Abstract A firm understanding of the biology of hematopoietic stem and progenitor cell (HSC/ HPC) trafficking is believed to be critical for the development of methodologies to improve transplant efficiency and subsequently immune reconstitution during hematopoietic stem cell transplantation in the clinical setting. Through the use of CD26 inhibitors and CD26 deficient mice (CD26−/−), we have previously generated data in mice suggesting that suppression of CD26/DPPIV (dipeptidylpeptidase IV) enzymatic activity on the transplant donor cell population can be utilized as a method of increasing transplant efficiency (Christopherson, KW 2nd, et al, Science 2004. 305:1000–3). However, the clinical importance of the transplant recipient should not to be overlooked given the potential importance of the bone marrow microenvironment in regulating the transplant process. We therefore investigated here whether inhibition or loss of CD26 activity in recipient mice would have an effect on transplant efficiency utilizing an in vivo congenic mouse model of transplantation. The short-term homing and long-term engraftment of BoyJ donor cells (expressing CD45.1+) into lethally irradiated control C57BL/6, CD26 inhibitor (Diprotin A) treated C57BL/6, or CD26−/− mice (expressing CD45.2+) was monitored by flow cytometric analysis of the bone marrow and peripheral blood at 24 hours and 6 months post-transplant respectively. Twenty-four hours post-transplant of 20×106 BoyJ mononuclear cells, we observed 8.85±0.58%, 10.69±1.01%, and 12.45±1.33% donor derived Sca-1+lin− cells in the bone marrow of recipient mice for control, Diprotin A treated, and CD26−/− recipient mice respectively. As compared to control mice, this represents a 20.8% increase (p=0.01) with CD26 inhibitor treatment and a 40.7% increase (p£0.05) resulting from the use of a CD26−/− recipient in short-term homing (N=5 mice per group). Six months post-transplant of 1×105 BoyJ mononuclear cells, we observed 39.90± 4.38%, 70.22± 3.72%, and 92.51± 1.04% donor contribution to hematopoiesis in the peripheral blood of control, Diprotin A treated, and CD26−/− recipient mice respectively. This represents a 76.0% increase (p£0.01) with CD26 inhibitor treatment and a 131.9% increase (p£0.01) as a result of the CD26−/− recipient in long-term engraftment as compared to control recipient mice (N=14 mice per group). These results provide pre-clinical evidence of the importance of CD26 expression within the transplant recipient with regard to regulating hematopoietic stem cell homing and engraftment. Our results also support the potential use of CD26 inhibitors to treat transplant patients during hematopoietic stem cell transplantation as a method of improving transplant efficiency. Lastly, our use of inhibitor treated C57BL/6 and CD26−/− recipient mice, which are also on a C57BL/6 background, in conjunction with a congenic model of transplantation provides a accurate and convenient model system for the in vivo testing of the efficacy of existing and new CD26 inhibitors in transplant recipients.


Author(s):  
Franco Aversa ◽  
Lucia Prezioso ◽  
Ilenia Manfra ◽  
Federica Galaverna ◽  
Angelica Spolzino ◽  
...  

The advantage of using a Human Leukocyte Antigen (HLA)-mismatched related donor is that almost every patient who does not have a HLA-identical donor or who urgently needs hematopoietic stem cell transplantation (HSCT) has at least one family member with whom shares one haplotype (haploidentical) and who is promptly available as a donor. The major challenge of haplo-HSCT is intense bi-directional alloreactivity leading to high incidences of graft rejection and graft-versus-host disease (GVHD). Advances in graft processing and in pharmacologic prophylaxis of GVHD have reduced these risks and have made haplo-HSCT a viable alternative for patients lacking a matched donor. Indeed, the haplo-HSCT  has spread to centers worldwide even though some centers have preferred an approach based on T cell depletion of G-CSF-mobilized peripheral blood progenitor cells (PBPCs), others have focused on new strategies for GvHD prevention, such as G-CSF priming of bone marrow and robust post-transplant immune suppression or post-transplant cyclophosphamide (PTCY). Today, the graft can be a megadose of T-cell depleted PBPCs or standard dose of unmanipulated bone marrow and/or PBPCs.  Although haplo-HSCT modalities are based mainly on high intensity conditioning regimens, recently introduced reduced intensity regimens (RIC)   showed promise in decreasing early transplant-related mortality (TRM), and extending the opportunity of HSCT to an elderly population with more comorbidities. Infections are still mostly responsible for toxicity and non-relapse mortality due to prolonged immunosuppression related, or not, to GVHD. Future challenges lie in determining the safest preparative conditioning regimen, minimizing GvHD and promoting rapid and more robust immune reconstitution.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1872-1872
Author(s):  
Zheng Hu ◽  
Feng Jin ◽  
Jin He ◽  
Chunhui Jin ◽  
Wei Fan ◽  
...  

Abstract Antithymocyte globulin (ATG) is often included in the conditioning regimen to prevent graft-vs.-host disease in allogeneic hematopoietic stem cell (HSC) transplantation. However, because ATG contains antibodies targeting a wide range of antigens on human cells, its potential off-target effects remain a concern. Here, we explored this question in humanized mice that were made by transplantation of human fetal thymic grafts (under the renal capsule) and CD34+ human fetal liver cells (i.v.). We showed that ATG binds to almost all lineages of human hematopoietic cells including HSCs, and accordingly, it is capable of depleting almost all human hematopoietic cells. Interestingly, the efficacy of ATG was highly variable depending on the tissue of residence, with human cells in bone marrow significantly less susceptible than those in the blood and spleen. Recovery of multilineage human lymphohematopoietic reconstitution in humanized mice that received ATG 3 weeks after HSC transplantation indicates that ATG had a minimal effect on human HSCs that have settled in bone marrow niches. However, efficient human HSC depletion and engraftment failure were seen in mice receiving ATG immediately following transplantation. Our data indicate that the efficacy of ATG is tissue-dependent, and suggest a risk of impairing donor hematopoietic engraftment when ATG is used in preparative conditioning regimens in HSC transplantation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 821-821 ◽  
Author(s):  
Marina Cavazzana-Calvo ◽  
Nathalie Cartier ◽  
Salima Hacein-Bey Abina ◽  
Gabor Veres ◽  
Manfred Schmidt ◽  
...  

Abstract We report preliminary results in 3 children with cerebral X-linked adrenoleukodystrophy (ALD) who received in September 2006, January 2007 and June 2008 lentiviral vector transduced autologous hematopoietic stem cell (HSC). We have previously demonstrated that cerebral demyelination associated with cerebral ALD can be stopped or reversed within 12–18 months by allogeneic HSC transplantation. The long term beneficial effects of HCT transplantation in ALD are due to the progressive turn-over of brain macrophages (microglia) derived from bone-marrow cells. For the current HSC gene therapy procedure, we used mobilized peripheral blood CD34+ cells that were transduced ex vivo for 18 hours with a non-replicative HIV1-derived lentiviral vector (CG1711 hALD) at MOI25 and expressing the ALD cDNA under the control of the MND (myeloproliferative sarcoma virus enhancer, negative control region deleted, dl587rev primer binding site substituted) promoter, and in the presence of 4 human recombinant cytokines (Il- 3, Stem Cell Factor [SCF], Flt3-ligand and Megakaryocyte Growth and Differentiation Factor [MGDF]) and CH-296 retronectine. Transduced cells were frozen to perform the required (RCL) safety tests. After thawing and prior to reinjection, 50%, 30% and 40% of transduced CD34+ cells expressed the ALD protein with a mean of 0.7, 0.6 and 0.65 copies of integrated provirus per cell. Transduced CD34+ cells were infused to ALD patients after a conditioning regimen including full doses of cyclophosphamide and busulfan. Hematopoietic recovery occured at day 13–15 post-transplant and the procedure was uneventful. In patient P1 and P2, the percentage of lymphocytes and monocytes expressing the ALD protein declined from day 60 to 6 months after gene therapy (GT) and remained stable up to 16 months post-GT. In P1, 9 to 13% of CD14+, CD3+, CD19+ and CD15+ cells expressed ALD protein 16 months post-transplant. In P2 and at the same time-point after transplant, 10 to 18% of CD14+, CD3+, CD19+ and CD15+ cells expressed ALD protein. ALD protein was expressed in 18–20% of bone marrow CD34+ cells from patients P1 and P2, 12 months post-transplant. In patient P3, 20 to 23% of CD3+, CD14+ and CD15+ cells expressed ALD protein 2 months after transplant. Tests assessing vector-derived RCL and vector mobilization were negative up to the last followups in the 3 patients. Integration of the vector was polyclonal and studies of integration sites arein progress. At 16 months post-transplant, HSC gene therapy resulted in neurological effects comparable with allogeneic HSC transplantation in patient P1 and P2. These results support that: ex-vivo HSC gene therapy using HIV1-derived lentiviral vector is not associated with the emergence of RCL and vector mobilization; a high percentage of hematopoietic progenitors were transduced expressing ALD protein in long term; no early evidence of selective advantage of the transduced ALD cells nor clonal expansion were observed. (This clinical trial is sponsored by Institut National de la Santé et de la Recherche Médicale and was conducted in part under a R&D collaboration with Cell Genesys, Inc., South San Francisco, CA)


Sign in / Sign up

Export Citation Format

Share Document