scholarly journals Genetic engineering of hoxb8 immortalized hematopoietic progenitors: a potent tool to study macrophage tissue migration

2019 ◽  
Author(s):  
Solene Accarias ◽  
Thibaut Sanchez ◽  
Arnaud Labrousse ◽  
Myriam Ben-Neji ◽  
Aurélien Boyance ◽  
...  

AbstractTumor-associated macrophages (TAM) are detrimental in most cancers. Controlling their recruitment is thus potentially therapeutic. We showed that TAM perform the protease-dependent mesenchymal migration in cancer, while macrophages perform amoeboid migration in other tissues. Inhibition of mesenchymal migration correlates with decreased TAM infiltration and tumor growth, providing rationale for a new cancer immunotherapy specifically targeting TAM motility. To identify new effectors of mesenchymal migration, we produced ER-Hoxb8-immortalized hematopoietic progenitors with unlimited proliferative ability. The functionality of macrophages differentiated from ER-Hoxb8 progenitors was compared to bone marrow-derived macrophages (BMDM). They polarized into M1- and M2-orientated macrophages, generated ROS, ingested particles, formed podosomes, degraded the extracellular matrix, adopted amoeboid and mesenchymal migration in 3D, and infiltrated tumor explants ex vivo using mesenchymal migration. We also used the CRISPR/Cas9 system to disrupt gene expression of a known effector of mesenchymal migration, WASP, to provide a proof of concept. We observed impaired podosome formation and mesenchymal migration capacity, thus recapitulating the phenotype of BMDM isolated from Wasp-KO mice. Thus, we validate the use of Hoxb8-macrophages as a potent tool to investigate macrophage functionalities.Summary statementWe validate the use of ER-Hoxb8-immortalized hematopoietic progenitors combined to CRISPR/Cas9 technology as a potent tool to investigate macrophage functionalities with a large scale of applications.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2358-2358
Author(s):  
Ali Nowrouzi ◽  
Africa Gonzales-Murillo ◽  
Anna Paruzynski ◽  
Ariana Jacome ◽  
Paula Rio ◽  
...  

Abstract Improved protocols using lentiviral vectors have been established with minimal cytokine exposure and short transduction times proving more suitable for overcoming the disease-specific challenge in correcting functionally defective hematopoietic stem cells (HSCs) of Fanconi Anemia (FA) patients. Bone marrow (BM) cells from FA patients were transduced ex vivo with lentiviral vectors (LVs) expressing FANCA and/or EGFP using optimized conditions to preserve the repopulating properties of the primitive hematopoietic stem cells (manuscript submitted). In a forward preclinical screening of possible LV-induced side effects we analyzed the insertional inventory in colonies generated by FA BM cells previously transduced with the LVs. We have established and optimized DNA and RNA isolation procedures for minimal cell numbers, suitable for large scale screening of colony forming cell (CFC) derived colonies by linear amplification-mediated PCR (LAM-PCR) and massive parallel pyrosequencing (454 GS Flx system; Roche). This approach is applicable for detecting early indicators of clonal selection, and is based on the analysis of common integration sites (CIS) and non-random distribution of vector insertions in particular genomic loci. From a total of 180 CFC-derived colonies expressing the EGFP LV marker gene, 298 vector insertions could be sequenced and mapped to the human genome. The analysis of vector targeted gene coding regions showed a non-random genomic distribution of LV insertions, with a significant overrepresentation of RefSeq genes that are part of distinct functional categories. Accordingly vector associated genes are predominantly involved in cellular signal cascades regulated by the MAP Kinase family known to be involved in a wide variety of cellular processes such as proliferation, differentiation, transcription regulation and development. Apart from the observed high integration frequency in genes (>80%), partial loss of vector LTR nucleotides was detected in >10% of the integrants (3–25bp). Notably, >20% of the lentiviral insertions were found to be located in CIS of predominantly 2nd order. Further screening assays of LV transduced CFC-derived colonies will allow a deeper investigation in the functional consequences of such CIS targeting in gene therapy protocols of FA. However our results suggest that the LV transduction of FA BM progenitors leads to a relatively high frequency of insertions in CIS which may be indicative of an insertion based (specific) selection mechanism. We herby show that the ex vivo large scale integration site analyses of CFC-derived colonies from patients considered to undergo gene therapeutic treatments constitutes a robust approach, which combined with mouse preclinical biosafety studies will help to improve the safety of clinical gene therapy protocols. The non-random distribution of LV integrations in CIS associated genes and in genes involved in particular cellular pathways may be indicative for the altered biochemical pathways characteristic of FA stem cells, with reported defects in DNA repair and self-renewal.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4775-4775
Author(s):  
Katharina Schallmoser ◽  
Christina Bartmann ◽  
Eva Rohde ◽  
Simone Bork ◽  
Christian Guelly ◽  
...  

Abstract Abstract 4775 Background: Based on promising experimental studies with mesenchymal stem and progenitor cells (MSPCs) multiple clinical trials have been initiated. In previous studies we have observed genomic stability of MSPCs after efficient short-term expansion in a humanized GMP compliant system with pooled human platelet lysate (pHPL) replacing fetal bovine serum (FBS) as the cell culture supplement (Schallmoser K. and Strunk D., Journal of Visualized Experiments (32) DOI: 10.3791/1523, 2009). Notably, depending on culture protocols, an extensive propagation with highly variable cell culture duration may be necessary to yield enough MSPCs for therapy. The decline in proliferation rates of MSPCs in the course of the different long-term expansion procedures may indicate a propensity for replicative senescence which may hamper long term functionality in vivo. We have therefore initiated a molecular profiling of senescence-associated regulated genes to determine the state of senescence before MSPC transplantation. Methods: Human bone marrow-derived MSPCs were cultured following a highly efficient two-passage protocol (primary culture of unseparated bone marrow and subsequent large scale expansion; Schallmoser K. et al., Tissue Engineering 14:185-196, 2008) compared to conventional serial passaging in three different growth conditions with regularly more then four passages to obtain comparable final cell numbers. Culture media were either supplemented with FBS in different concentrations or pHPL. Gene expression changes were tested by microarray analysis and selected targets were reanalyzed by quantitative real-time PCR. The genomic stability of MSPCs after long-term culture was determined by array comparative genomic hybridization (CGH). Results: Despite high proliferation rate large scale expanded MSPCs showed genomic stability in array CGH. Long-term MSPC growth induced similar gene expression changes in MSPCs irrespective of isolation and expansion conditions. In particular, genes involved in cell differentiation, apoptosis and cell death were up-regulated, whereas genes involved in mitosis and proliferation were down-regulated. Furthermore, overlapping senescence-associated gene expression changes were found in all MSPC preparations. The genomic copy number variations detected in MSPCs of early and late passages in all culture conditions did not coincide with differentially expressed genes. Conclusion: Our data indicate that MSPC expansion can induce gene expression changes independent of isolation and FBS-supplemented as well as FBS-free expansion conditions. A panel of genes will be presented that might offer a practicable approach to assess MSPC quality with regard to the state of replicative senescence in advance of therapeutic application. Determining the impact of senescence acquired during cell expansion on the therapeutic potential of MSCPs for both immune modulation and organ regeneration may help to develop more efficient treatment strategies. Disclosures: No relevant conflicts of interest to declare.


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.


Author(s):  
Atefeh Soltani ◽  
Saeid Abroun ◽  
Mojtaba Rezazadeh Valojerdi ◽  
Bahareh Vahidianfar ◽  
Elahe Sadat Hosseini

Background and Aims: Bone marrow-derived mesenchymal stem cells (BM-MSCs) are a well-known source of multipotent adult stem cells. Despite using different methodologies of MSCs preparing for clinical applications, the top safest procedure to manipulate these cells, has not yet been determined. Recently, ex-vivo expansion of MSCs for their subsequent implantation, using some biological product, is suggested instead of fetal bovine serum (FBS). Previous studies have shown the effect of follicular fluid (FF) (a dynamic fluid in ovarian follicle) as an additive component in cell culture. Hence, this study aimed to decipher its role on the human BM-MSC proliferation.Materials and Methods: In this study, BM-MSCs at 3rd passage were cultivated in the presence of 20% FF (group I), 10% FF+ FBS 10% (group II) and FBS 20% as control group. The capacity of proliferation as calculating population doubling times and gene expression levels of stem cell factor, stromal cell-derived factor 1, and transforming growth factor beta were analyzed in osteogeneic media to examine the impacts of FF on osteogenesis of MSCs.Results: Our results corroborated an up-regulatory effect of FF on the proliferation of BM-MSCs by shorter population doubling times in the group II of treated cells and an increase in gene expression level of osteocalcin and transforming growth factor beta in the presence of higher concentrations of FF in cell culture  FF 20% and 10%, respectively.Conclusions: FF is a potent mitogen for cell proliferation. FF may be an efficient substitution of FBS in ex-vivo cell culture, eliminating zoonotic infections and immunological reactions.


2010 ◽  
Vol 69 (10) ◽  
pp. 1880-1885 ◽  
Author(s):  
J. R. Lamas ◽  
L. Rodriguez-Rodriguez ◽  
A. G. Vigo ◽  
R. Alvarez-Lafuente ◽  
P. Lopez-Romero ◽  
...  

Cancers ◽  
2021 ◽  
Vol 13 (17) ◽  
pp. 4370
Author(s):  
Rafael Pelaez ◽  
Rodrigo Ochoa ◽  
Ana Pariente ◽  
Angela Villanueva-Martínez ◽  
Álvaro Pérez-Sala ◽  
...  

Sterculic acid (SA) is a cyclopropenoid fatty acid isolated from Sterculia foetida seeds. This molecule is a well-known inhibitor of SCD1 enzyme, also known as ∆9-desaturase, which main function is related to lipid metabolism. However, recent studies have demonstrated that it also modifies many other pathways and the underlying gene expression. SCD overexpression, or up-regulated activity, has been associated with tumor aggressiveness and poor prognosis in many cancer types. Scd1 down-regulation, with different inhibitors or molecular strategies, reduces tumor cell survival and cell proliferation, as well as the chemoresistance associated with cancer stem cell presence. However, SA effects over cancer cell migration and extracellular matrix or adhesion molecules have not been described in cancer cells up to now. We used different migration assays and qPCR gene expression analysis to evaluate the effects of SA treatment in cancer cells. The results reveal that SA induces tumoral cell death at high doses, but we also observed that lower SA-treatments induce cell adhesion-migration capacity reduction as a result of modifications in the expression of genes related to integrins and extracellular matrix compounds. Overall, the functional and transcriptomic findings suggest that SA could represent a new inhibitor activity of epithelial to mesenchymal transition.


2020 ◽  
Author(s):  
Eva Camarillo-Retamosa ◽  
Luke Watson ◽  
Paul Loftus ◽  
Senthil Alagesan ◽  
Yvonne O’Donoghue ◽  
...  

AbstractThe marrow microenvironment is a complex and heterogeneous mixture of hematopoietic and stromal progenitors necessary for haematopoiesis. Whilst the hematopoietic progenitors are well described, the stromal cellular composition is not fully elucidated due to the low cells numbers, localisation-distribution-accessibility, and the lack of specific biomarkers. Cellular taxonomy studies have recently identified new populations of stromal subsets with distinct gene signature and regulatory properties of hematopoietic regeneration. Fibroblast activation protein-α (FAP), a stromal cell type first identified in cancer is also rarely found in normal tissues but might play an essential role in tissue homeostasis. Using FAPDM2 transgenic mouse in which FAP-expressing cells can be ablated with Diphtheria Toxin (DTX) FAP+ cells were depleted in healthy mice. Whilst FAP+ cells constituted 5% of all marrow cells; its ablation caused a rapid loss of PDGFR-α, Leptin-R, gp38 and SDC2 stromal cells populations, endothelial cells and vascular disruption. These resulted in anaemia, thrombocytopenia and neutropenia in peripheral blood (PB) and extreme hypo-cellularity in marrow with abnormalities within the hematopoietic progenitors. In an effort to reverse the phenotypes caused by FAP+ cell loss, a single intravenous injection of syngeneic bone marrow-derived stromal cells was administered. In a short-term evaluation, anaemia, thrombocytopenia and neutropenia ameliorated in PB and the numbers of marrow hematopoietic progenitors increased. Our data suggest FAP-expressing cells are a non-redundant component of the marrow microenvironment, necessary for marrow homeostasis and haematopoiesis. These data also provided evidence that stromal cell ablation can be rescued by stromal cell therapy.Significance StatementFAP-expressing cells depletion led to collateral damage in PB and marrow, including haematological defects that can be ameliorated by adoptive transfer of low-dose, ex-vivo expanded FAP-expressing marrow stromal cells. We suggest that stromal cell loss is a feature of severe immune-mediated inflammatory diseases – such as Graft versus Host Disease and sepsis - and that FAPDM2 model represents a novel tool to explore the native function of the recently identified stromal cell sub-populations.


2016 ◽  
Vol 136 (5) ◽  
pp. S69
Author(s):  
P.P. Soldati ◽  
F.M. Thomaz ◽  
F. Paes ◽  
S. Moura ◽  
P. Moncayo ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2303-2303
Author(s):  
M. Kozik ◽  
J. J. Banks ◽  
L. R. Fanning ◽  
M. R. Finney ◽  
Y. Huang ◽  
...  

Abstract Background. Umbilical cord blood (UCB), a source of hematopoeitic stem cells (HSC), is marked by delayed engraftment attributed to the limited cellular content of a single UCB unit. Cytokine-based ex vivo expansion of UCB is a way of increasing the number of cells available for allogeneic transplants, however, this strategy has not demonstrated improved engraftment in human clinical trials. Further studies have incorporated human mesenchymal stem cells (huMSC) which may provide signals that control the proliferation, survival, and differentiation of HSC. In attempt to reduce the occurrence and severity of GVHD following allogeneic transplants, strategies such as utilizing T-cell depleted grafts have been pursued, however, clinical trials using these grafts have shown decreased rates of donor engraftment, suggesting the requirement of accessory cells as well as HSC to achieve engraftment. Methods. UCB mononuclear cells (MNC) were cultured using cytokines (IL-3, IL-6, G-CSF, SCF, Flt-3L, EPO) with or without a feeder-layer of huMSC for 12 days. On day 12, viability, 4-color flow cytometry, and human engraftment potential were measured. Human engraftment potential was determined by injecting cells (without CD34+ selection) from each culture condition and non-cultured UCB MNC, via tail vein, into sublethal irradiated NOD/SCID mice. Mice were injected with unexpanded UCB MNC (n=23), UCB expanded in huMSC+cytokines (n=21) and UCB expanded in cytokines alone (n=10). 7–9 weeks following injection of human cells, bone marrow was harvested and analyzed for human content. Positive human engraftment was determined by a human %CD45+ of ≥ 0.4%. Results. An 8.77 fold expansion of UCB cultured in cytokines alone compared to a 7.14 fold expansion of UCB cultured in huMSC + cytokines was observed. Surface phenotyping of expanded UCB, and human cells emerging in the bone marrow of NOD/SCID mice following injection of cultured and non-cultured UCB are in Table 1. Unexpanded huMSC+cytokines Cytokines % CD3 44.0 (4.40M) 2.04 (10.1M) 1.52 (4.35M) % CD56 17.0 (1.70M) 7.07 (36.1M) 3.69 (13.8M) % CD34 3.41 (.341M) 2.25 (10.9M) 3.52 (12.3M) Bone marrow of NOD/SCID mice % CD45+ 2.57 3.58 2.38 % of CD45+ co-expressing CD3 11.0 9.25 18.7 % of CD45+ co-expressing CD19 33.5 16.6 19.4 % of CD45+ co-expressing CD56 10.1 8.04 1.60 Human engraftment was seen in 13 mice which received unexpanded UCB, 10 mice which received UCB expanded in huMSC+cytokines and only 3 mice which received UCB expanded in cytokines alone. Statistical analysis, using multivariable logistic regression to determine the factors that predict engraftment, revealed that the proportions of T and NK cells present in expanded UCB correlated with engraftment. A 10% increase in the proportion of CD45+ co-expressing CD3 was associated with a 1.79 fold increase in engraftment (p=0.016), whereas each 10% increase in the proportion of CD45+ co-expressing CD56 increased the odds of engrafting by 104% (p= 0.003). Conclusions. We observed an expansion of CD34 hematopoietic progenitors as well as a greater proportion of CD3+ cells, in expansion conditions incorporating huMSC. Additionally, we observed improved rates of engraftment in this expansion condition. Therefore, although the mechanism by which accessory cells including T and NK cells facilitate HSC engraftment is not known, we observed that the presence of accessory cells in addition to CD34 hematopoietic progenitors facilitated engraftment in NOD/SCID mice.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4743-4743
Author(s):  
Marijke W. Maijenburg ◽  
Willy A. Noort ◽  
Marion Kleijer ◽  
Charlotte J.A. Kompier ◽  
Kees Weijer ◽  
...  

Abstract It is thought that adult mesenchymal stromal cells (MSC) are important for tissue repair and maintenance. Crucial in these processes is the presence of MSC at the site of injury, however the recruitment and migration of MSC towards their destiny is poorly understood. With respect to future cell therapy, we are studying the process of migration of various human mesenchymal stem cell sources, and hypothesize that only a subpopulation of ex vivo expanded mesenchymal stem cells is capable of specific homing. For this purpose, MSC from different sources i.e. fetal lung (FL), fetal bone marrow (FBM), adult bone marrow (ABM) and adult adipose tissue (AT) were derived by plastic adherence and subsequently expanded. All MSC sources were characterized as CD73+, CD90+, CD105+, CD34− and CD45−. MSC (P4-9) were allowed to migrate for 4h towards SDF-1a, PDGF-BB, HGF, bFGF or FCS over fibronectin-coated 12 mm pore size transwell plates. FL-MSC migrated significantly better towards SDF-1a as compared to ABM-MSC or AT-MSC. This enhanced migration capacity towards SDF-1a is specific for FL-MSC since AT-MSC migrated better towards FCS as compared to FL-MSC. Even ABM-MSC responded better to FCS than FL-MSC. This suggests that MSC originating from all sources are able to migrate but require different triggers to induce migration. In order to elucidate whether the observed differences in migration potential were due to developmental stage, cultured MSC derived from fetal bone marrow were tested as well. No significant differences in migration capacity were observed between adult and fetal BM- MSC for any of the (chemotactic) stimuli evaluated. Interestingly, FL-MSC had a significant increased migration capacity as compared to FBM-MSC towards SDF-1a, PDGF-BB and HGF, suggesting that the origin of tissue may determine migration capacity of ex vivo expanded MSC. Since it was observed that only a small percentage of the cultured MSC were able to migrate towards the various stimuli, checkerboard migration was performed to elucidate whether a synergistic effect could be observed. No synergistic effect was observed between SDF and PDGF, SDF and FCS or PDGF and FCS in FL-MSC, suggesting that there may be one subpopulation of MSC that possesses migratory capacities. When studying the SDF-1-induced migratory subpopulation in more detail, it was observed that, after migration, migratory MSC originating from all tissue sources maintain their proliferation and differentiation capacity and express CXCR4 at a higher level than MSC that did not migrate. To be able to migrate, cells have to rearrange their actin cytoskeleton and focal adhesions. These processes can be initiated by various chemokines and growth factors. In response to SDF or FCS, morphological changes were observed in ABM-MSC by confocal microscopy. Cells became smaller and membrane protrusions appeared, whereas this was absent in the control. Furthermore, upon stimulation with SDF, PDGF and FCS, tyrosine-phosphorylation of the adapter protein paxillin that links the actin cytoskeleton to focal adhesions was increased. In conclusion, our results suggest that migration potential of ex vivo expanded MSC derived from various adult and fetal tissues have different migratory capacity towards growth factor and chemokine stimuli and may involve paxillin phosphorylation. Our data indicate that further studies on the migratory subpopulation(s) within the heterogeneous population of culture expanded MSC will contribute to unravel how and which MSC will be of interest for future cellular therapies.


Sign in / Sign up

Export Citation Format

Share Document