scholarly journals TGFβ Signaling in Myeloid Cells Promotes Lung and Liver Metastasis Through Different Mechanisms

2021 ◽  
Vol 11 ◽  
Author(s):  
Cristina Stefanescu ◽  
Merel Van Gogh ◽  
Marko Roblek ◽  
Mathias Heikenwalder ◽  
Lubor Borsig

TGFβ overexpression is commonly detected in cancer patients and correlates with poor prognosis and metastasis. Cancer progression is often associated with an enhanced recruitment of myeloid-derived cells to the tumor microenvironment. Here we show that functional TGFβ-signaling in myeloid cells is required for metastasis to the lungs and the liver. Myeloid-specific deletion of Tgfbr2 resulted in reduced spontaneous lung metastasis, which was associated with a reduction of proinflammatory cytokines in the metastatic microenvironment. Notably, CD8+ T cell depletion in myeloid-specific Tgfbr2-deficient mice rescued lung metastasis. Myeloid-specific Tgfbr2-deficiency resulted in reduced liver metastasis with an almost complete absence of myeloid cells within metastatic foci. On contrary, an accumulation of Tgfβ-responsive myeloid cells was associated with an increased recruitment of monocytes and granulocytes and higher proinflammatory cytokine levels in control mice. Monocytic cells isolated from metastatic livers of Tgfbr2-deficient mice showed increased polarization towards the M1 phenotype, Tnfα and Il-1β expression, reduced levels of M2 markers and reduced production of chemokines responsible for myeloid-cell recruitment. No significant differences in Tgfβ levels were observed at metastatic sites of any model. These data demonstrate that Tgfβ signaling in monocytic myeloid cells suppresses CD8+ T cell activity during lung metastasis, while these cells actively contribute to tumor growth during liver metastasis. Thus, myeloid cells modulate metastasis through different mechanisms in a tissue-specific manner.

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 734-734
Author(s):  
Miriam Y Kim ◽  
Matthew L Cooper ◽  
Julie K Ritchey ◽  
Julia Hollaway ◽  
John F. DiPersio

Abstract Chimeric antigen receptor (CAR) T cells are effective against B cell malignancies and multiple myeloma, but their efficacy has been limited to date for acute myeloid leukemia (AML). We sought to investigate whether there were fundamental differences in targeting B cell antigens as compared to myeloid antigens with CAR T cells, that may shed light on the mechanism of CAR T cell resistance in patients with AML. For these studies, we utilized human CAR T cells targeting CD19 (CART19) and CD33 (CART33), canonical B cell and myeloid cell antigens, respectively. To ensure that the potency of the two CAR constructs were equivalent, we generated dual CD19 and CD33 expressing cell lines, by adding CD33 to Ramos, a CD19+ B lymphoblastic cell line, and adding CD19 to THP-1, a CD33+ myeloid cell line. We confirmed that CART19 and CART33 were equally potent against CD33+Ramos and CD19+THP-1 cells. To investigate the influence of normal hematopoietic cells on CAR T cell behavior, we incubated CD19+THP-1 cells with CART19 and CART33 in the presence of peripheral blood (PB) or bone marrow (BM) mononuclear cells. We found that both PB and BM enhanced tumor clearance to a similar degree for each CAR construct. Additionally, IL-6 was detected in the supernatant of PB or BM co-cultured with CART19 and CART33, and these levels were markedly increased in the presence of tumor cells. Notably, THP-1 cells by themselves produced high levels of IL-6 upon exposure to CAR T cells, likely reflecting the myeloid origin of this cell line, while Ramos cultured with these same CAR T cells did not produce IL-6. We assessed other myeloid cell lines (U937, KG-1, Kasumi-3, Molm13, HL-60, and K562) and also noted IL-6 production when co-cultured with CART33, although the levels were significantly lower than that produced by THP-1. Of note, IL-6 levels were slightly but consistently higher with CART19 than with CART33 in these in vitro assays, which we attribute to the loss of normal myeloid cells from CART33-mediated killing. To study the effects of normal hematopoiesis on human CAR T cell activity in vivo, we injected NSGS mice with human cord blood CD34+ hematopoietic stem cells (HSCs) to generate a human hematopoietic system in these mice, followed by administration of untransduced (UTD) control T cells, CART19 or CART33. To prevent the confounding effect of allogeneic killing, CAR T cells were generated from T cells of the same cord blood product as the CD34+ cells. We confirmed the expected loss of human CD19+ B cells and CD33+ myeloid cells in the peripheral blood after CART19 and CART33 treatment, respectively. Surprisingly, we found that only CART33 treatment led to elevated plasma human IL-6 levels in this model. We then injected CD19+THP-1 cells to the mice after HSC engraftment, to assess the anti-tumor activity of the CAR T cells and to increase the potential for toxicity. Consistent with our in vitro data, mice with a human hematopoietic system cleared tumor more efficiently than mice without prior HSC engraftment after treatment with CART19 or CART33. However, while we observed mild weight loss and IL-6 elevation in mice after CART19 treatment, this effect was much more pronounced in mice that received CART33. We hypothesized that the presence of antigen on normal myeloid cells both increased the toxicity and decreased the efficacy of CART33, due to a massive release of inflammatory cytokines from myeloid cells in the immediate aftermath of CART33 treatment, followed by loss of the augmentation of CAR T cell activity mediated by myeloid cells in the long term. To test this hypothesis, we engrafted mice with either control HSCs or CD33 KO HSCs, followed by injection of THP-1 and CART33. Only mice with CD33 KO HSCs maintained myeloid cells after CART33, as expected. CD33 KO HSC-engrafted mice exhibited less toxicity after CART33 treatment than mice with control HSCs, in that they did not lose weight or demonstrate elevated IL-6 levels. Furthermore, absence of CD33 on myeloid cells led to enhanced CAR T cell expansion and persistence, that resulted in better long-term tumor control. In summary, our data suggests that targeting myeloid antigens with CAR T cells may be intrinsically self-defeating due to loss of myeloid cells that are required for sustained CAR T cell activity. These studies illuminate the challenges when extending CAR T cell therapy to myeloid malignancies, and highlight the importance of normal myeloid cells in augmenting T cell-based immunotherapies. Figure 1 Figure 1. Disclosures Kim: Tmunity: Patents & Royalties; NeoImmune Tech: Patents & Royalties. Cooper: RiverVest: Consultancy; Wugen: Current Employment, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company, Patents & Royalties; NeoImmune Tech: Patents & Royalties.


2020 ◽  
Vol 5 (43) ◽  
pp. eaay1863 ◽  
Author(s):  
Laura Strauss ◽  
Mohamed A. A. Mahmoud ◽  
Jessica D. Weaver ◽  
Natalia M. Tijaro-Ovalle ◽  
Anthos Christofides ◽  
...  

PD-1, a T cell checkpoint receptor and target of cancer immunotherapy, is also expressed on myeloid cells. The role of myeloid-specific versus T cell–specific PD-1 ablation on antitumor immunity has remained unclear because most studies have used either PD-1–blocking antibodies or complete PD-1 KO mice. We generated a conditional allele, which allowed myeloid-specific (PD-1f/fLysMcre) or T cell–specific (PD-1f/fCD4cre) targeting of Pdcd1 gene. Compared with T cell–specific PD-1 ablation, myeloid cell–specific PD-1 ablation more effectively decreased tumor growth. We found that granulocyte/macrophage progenitors (GMPs), which accumulate during cancer-driven emergency myelopoiesis and give rise to myeloid-derived suppressor cells (MDSCs), express PD-1. In tumor-bearing PD-1f/fLysMcre but not PD-1f/fCD4cre mice, accumulation of GMP and MDSC was prevented, whereas systemic output of effector myeloid cells was increased. Myeloid cell–specific PD-1 ablation induced an increase of T effector memory cells with improved functionality and mediated antitumor protection despite preserved PD-1 expression in T cells. In PD-1–deficient myeloid progenitors, growth factors driving emergency myelopoiesis induced increased metabolic intermediates of glycolysis, pentose phosphate pathway, and TCA cycle but, most prominently, elevated cholesterol. Because cholesterol is required for differentiation of inflammatory macrophages and DC and promotes antigen-presenting function, our findings indicate that metabolic reprogramming of emergency myelopoiesis and differentiation of effector myeloid cells might be a key mechanism of antitumor immunity mediated by PD-1 blockade.


2020 ◽  
Vol 8 (Suppl 2) ◽  
pp. A59.2-A60
Author(s):  
A Osipov ◽  
L Zheng

BackgroundPancreatic ductal adenocarcinoma (PDAC) is resistant to immune checkpoint inhibition. One of the major resistance mechanisms is attributed to myeloid cells as an immunosuppressive element within the stroma of PDAC. It has been reported that focal adhesion kinase inhibitor (FAKi) can suppress immunosuppressive myeloid cells such as tumor associated macrophages (TAMs) and myeloid derived suppressor cells (MDSC), consequently sensitizing tumor to anti-PD1 antibody in mouse models of PDAC. Our group has previously shown in a murine model that targeting the stroma via PEGylated recombinant human hyaluronidase (PEGPH20) enhanced the anti-tumor activity of the whole cell vaccine (GVAX) by targeting CXCR4-expressing myeloid cells and led to an increase in infiltration of CCR7- effector memory T cell subsets. Here, we evaluate the hypothesis that FAK expressing myeloid cell subsets modulate T cell infiltration in human PDAC and FAKi can synergize with PEGPH20 by targeting myeloid cells in PDAC.Material and MethodsResected human PDAC tissue specimens treated with GVAX and anti-PD1 therapy was used to assess FAK expression in myeloid cell subsets and its impact on T cell infiltration. A sequential staining and stripping multiplex IHC technique that incorporates 28 myeloid and lymphoid biomarkers, as well as phosphorylated FAK (pFAK) combined with computational image processing was used to assess myeloid cell populations, T cell infiltration and FAK expression.An established murine model of metastatic PDAC treated with and without anti-PD1 therapy was used to assess the synergy and immune-modulating effect of FAKi and stromal degradation of hyaluronan via PEGPH20.ResultsIn human PDAC, FAK is widely expressed in TAMs and neutrophils. Increased FAK expression is associated with increased CXCR4 expression. Lower pFAK density in neutrophils and M2 TAMs, but not lower pFAK density in M1 TAMs, is associated with higher CD8+ T cell infiltration.FAKi and combination of FAKi with anti-PD1 extends survival in the mouse metastasis model of PDAC. Adding PEGPH20 to FAKi and anti-PD1 antibody significantly prolonged survival in this model. Comparing to the combination of FAKi and anti-PD1 antibody, adding PEGPH20 significantly decreased the number of CXCR4-expressing myeloid cells in the tumor microenvironment (TME) of PDAC and consequently led to an increase in the amount of CCR7+ central memory T cells. Additionally, the amount of G-MDSCs, inflammatory resident monocytes and PDL1 expressing myeloid cells in the TME of PDAC, was also decreased in PDAC treated with the triple combination of PEGPH20, FAKi and anti-PD1 antibody compared to FAKi and anti-PD1 antibody.ConclusionFAK is widely expressed in myeloid cell populations, directly correlated with CXCR4 expression and decreased FAK expression in a myeloid (M2 TAMs, neutrophil) inflamed stroma is associated with infiltration of effector CD8 T cells in human PDAC. Stromal degradation of hyaluronan via PEGPH20 combined with FAKi and anti-PD1 antibody further depletes immunosuppressive cells in the TME including G-MDSCs, inflammatory resident monocytes and PDL1 expressing myeloid cells and appears to target the CXCR4 pathway through PEGPH20. These findings support testing the combination of FAKi and anti-PD1 antibody with agents targeting CXCR4 directly or indirectly by PEGPH20 in human PDAC.Disclosure InformationA. Osipov: None. L. Zheng: None.


2016 ◽  
Vol 8 (334) ◽  
pp. 334ra52-334ra52 ◽  
Author(s):  
Marij J. Welters ◽  
Tetje C. van der Sluis ◽  
Hélène van Meir ◽  
Nikki M. Loof ◽  
Vanessa J. van Ham ◽  
...  

Therapeutic vaccination with human papillomavirus type 16 synthetic long peptides (HPV16-SLPs) results in T cell–mediated regression of HPV16-induced premalignant lesions but fails to install clinically effective immunity in patients with HPV16-positive cervical cancer. We explored whether HPV16-SLP vaccination can be combined with standard carboplatin and paclitaxel chemotherapy to improve immunity and which time point would be optimal for vaccination. This was studied in the HPV16 E6/E7–positive TC-1 mouse tumor model and in patients with advanced cervical cancer. In mice and patients, the presence of a progressing tumor was associated with abnormal frequencies of circulating myeloid cells. Treatment of TC-1–bearing mice with chemotherapy and therapeutic vaccination resulted in superior survival and was directly related to a chemotherapy-mediated altered composition of the myeloid cell population in the blood and tumor. Chemotherapy had no effect on tumor-specific T cell responses. In advanced cervical cancer patients, carboplatin-paclitaxel also normalized the abnormal numbers of circulating myeloid cells, and this was associated with increased T cell reactivity to recall antigens. The effect was most pronounced starting 2 weeks after the second cycle of chemotherapy, providing an optimal immunological window for vaccination. This was validated with a single dose of HPV16-SLP vaccine given in this time window. The resulting proliferative HPV16-specific T cell responses were unusually strong and were retained after all cycles of chemotherapy. In conclusion, carboplatin-paclitaxel therapy fosters vigorous vaccine-induced T cell responses when vaccination is given after chemotherapy and has reset the tumor-induced abnormal myeloid cell composition to normal values.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 374-374 ◽  
Author(s):  
Zhong-fa Yang ◽  
Karen Drumea ◽  
Alan G. Rosmarin

Abstract GABP is an ets transcription factor that regulates genes that are required for innate immunity, including CD18 (β2 leukocyte integrin), lysozyme, and neutrophil elastase. GABP consists of two distinct and unrelated proteins. GABPα binds to DNA through its ets domain and recruits GABPβ, which contains the transactivation domain; together, they form a functional tetrameric transcription factor complex. We recently showed that GABP is required for entry into S phase of the cell cycle through its regulation of genes that are required for DNA synthesis and cyclin dependent kinase inhibitors (Yang, et al. Nature Cell Biol9:339, 2007). Furthermore, GABP is an essential component of a retinoic acid responsive myeloid enhanceosome (Resendes and Rosmarin Mol Cell Biol26:3060, 2006). We cloned Gabpa (the gene that encodes mouse Gabpα) from a mouse genomic BAC library and prepared a targeting vector in which the ets domain is flanked by loxP recombination sites (floxed allele). Deletion of both floxed Gabpa alleles causes an early embryonic lethal defect. In order to define the role of Gabpα in myelopoiesis, we bred floxed Gabpa mice to mice that bear the Mx1-Cre transgene, which drives expression of Cre recombinase in response to injection of the synthetic polynucleotide, poly I-C. Deletion of Gabpa dramatically reduced granulocytes and monocytes in the peripheral blood, spleen, and bone marrow, but myeloid cells recovered within weeks. In vitro colony forming assays indicated that myeloid cells in these mice were derived only from Gabpa replete myeloid precursors (that failed to delete both Gabpa alleles), suggesting strong pressure to retain Gabpα in vivo. We used a novel competitive bone marrow transplantation approach to determine if Gabp is required for myeloid cell development in vivo. Sub-lethally irradiated wild-type recipient mice bearing leukocyte marker CD45.1 received equal proportions of bone marrow from wild type CD45.1 donor mice and floxed-Mx1-Cre donor mice that bear CD45.2. Both the CD45.2 (floxed-Mx1-Cre) and CD45.1 (wild type) bone marrow engrafted well. Mice were then injected with pI-pC to induce Cre-mediated deletion of floxed Gabpa. The mature myeloid and T cell compartments were derived almost entirely from wild type CD45.1 cells. This indicates that the proliferation and/or differentiation of myeloid and T cell lineages requires Gabp. In contrast, B cell development was not impaired. We conclude that Gabpa disruption causes a striking loss of myeloid cells in vivo and corroborates prior in vitro data that GABP plays a crucial role in proliferation of myeloid progenitor cells.


2021 ◽  
Vol 12 ◽  
Author(s):  
Milos Gojkovic ◽  
Pedro P. Cunha ◽  
Gabriella S. Darmasaputra ◽  
Laura Barbieri ◽  
Helene Rundqvist ◽  
...  

Myeloid cell interactions with cells of the adaptive immune system are an essential aspect of immunity. A key aspect of that interrelationship is its modulation by the microenvironment. Oxygen is known to influence myelosuppression of T cell activation in part via the Hypoxia inducible (HIF) transcription factors. A number of drugs that act on the HIF pathway are currently in clinical use and it is important to evaluate how they act on immune cell function as part of a better understanding of how they will influence patient outcomes. We show here that increased activation of the HIF pathway, either through deletion of the negative regulator of HIF, the von Hippel-Lindau (VHL) gene, in myeloid cells, or through pharmacological inhibitors of VHL-mediated degradation of HIF, potently suppresses T cell proliferation in myeloid cell/T cell culture. These data demonstrate that both pharmacological and genetic activation of HIF in myeloid cells can suppress adaptive cell immune response.


Blood ◽  
2000 ◽  
Vol 95 (11) ◽  
pp. 3349-3356 ◽  
Author(s):  
Takeshi Kuwata ◽  
I-Ming Wang ◽  
Tomohiko Tamura ◽  
Roshini M. Ponnamperuma ◽  
Rachel Levine ◽  
...  

Abstract To examine the role of retinoids in hematopoietic cell growth in vivo, we studied female SENCAR mice made vitamin A deficient by dietary restriction. Deficient mice exhibited a dramatic increase in myeloid cells in bone marrow, spleen, and peripheral blood. The abnormal expansion of myeloid cells was detected from an early stage of vitamin A deficiency and contrasted with essentially normal profiles of T and B lymphocytes. This abnormality was reversed on addition of retinoic acid to the vitamin A–deficient diet, indicating that the myeloid cell expansion is a direct result of retinoic acid deficiency. TUNEL analysis indicated that spontaneous apoptosis, a normal process in the life cycle of myeloid cells, was impaired in vitamin A–deficient mice, which may play a role in the increased myeloid cell population. Quantitative reverse transcriptase-polymerase chain reaction analysis of purified granulocytes showed that expression of not only RAR, but RXRs, 2 nuclear receptors that mediate biologic activities of retinoids, was significantly reduced in cells of deficient mice. This work shows that retinoids critically control the homeostasis of myeloid cell population in vivo and suggests that deficiency in this signaling pathway may contribute to various myeloproliferative disorders.


2020 ◽  
Vol 79 (11) ◽  
pp. 1453-1459 ◽  
Author(s):  
Andrey Kruglov ◽  
Marina Drutskaya ◽  
Dirk Schlienz ◽  
Ekaterina Gorshkova ◽  
Katharina Kurz ◽  
...  

ObjectivesNeutralisation of tumour necrosis factor (TNF) is widely used as a therapy for rheumatoid arthritis (RA). However, this therapy is only effective in less than a half of patients and is associated with several side effects. We hypothesised that TNF may possess non-redundant protective and immunomodulatory functions in vivo that cannot be blocked without a cost. The present work aimed to identify cellular sources of protective and pathogenic TNF, and its molecular forms during autoimmune arthritis.MethodsMice lacking TNF expression by distinct cell types, such as myeloid cells and T or B lymphocytes, were subjected to collagen-induced arthritis (CIA) and collagen antibody-induced arthritis. Mice lacking soluble TNF production were also employed. The severity and incidence of the disease, as well as humoral and cellular responses were assessed.ResultsMyeloid cell-derived TNF contributes to both induction and pathogenesis of autoimmune arthritis. Conversely, T cell-derived TNF is protective during the induction phase of arthritis via limiting of interleukin-12 production by dendritic cells and by subsequent control of autoreactive memory T cell development, but is dispensable during the effector phase of arthritis. B cell-derived TNF mediates severity of CIA via control of pathogenic autoantibody production.ConclusionsDistinct TNF-producing cell types may modulate disease development through different mechanisms, suggesting that in arthritis TNF ablation from restricted cellular sources, such as myeloid cells, while preserving protective TNF functions from other cell types may be superior to pan-anti-TNF therapy.


2015 ◽  
Vol 33 (3_suppl) ◽  
pp. 624-624 ◽  
Author(s):  
Julie G. Grossman ◽  
Timothy M. Nywening ◽  
Brian Belt ◽  
Elizabeth Pittman ◽  
Andrew Giorgi ◽  
...  

624 Background: Colorectal cancer (CRC) is the most common gastrointestinal malignancy. 60% of CRC patients are diagnosed with metastatic (m) CRC and the 5-year survival is <20%. Myeloid cells, particularly inflammatory monocytes (IM), are recruited from the bone marrow to the tumor microenvironment where they become tumor associated macrophages and play a crucial role in tumor progression, metastasis, and chemoresistance. While the importance of IM have been shown in other malignancies, little is known about their role in human CRC. Methods: Human tissue was collected under an IRB approved protocol at Washington University. Flow cytometry was performed on PBMCs and single cell suspensions of normal tissue and tumor samples. Qualitative RT-PCR and confocal microscopy were performed for CCL2. T-cell suppression assays were performed using CD14+ IM isolated from patient peripheral blood and tumor samples. Results: Analysis of pre-operative blood revealed that monocyte levels correlate with the extent of disease burden. Monocytes were elevated in CRC patients compared with controls (p<0.0001), additionally patients with liver metastasis had further elevation in monocytes compared with patient’s with primary disease (p=0.01). In metastatic patients, monocyte levels also correlate with survival following resection of hepatic metastasis (p=0.0002). FACS analysis confirmed these findings and demonstrated that the circulating CD11b+/CD14+/CCR2+ subset of IM was responsible for the increase. Both primary CRC and liver mCRC had increased expression of CCL2 compared to uninvolved tissue (p=0.008 and p=0.03, respectively). Production of CCL2 was localized to CRC cells. FACS analysis showed CCR2+ tumor infiltrating macrophages were elevated in CRC liver metastasis compared to adjacent normal liver and a paucity of effector T-cells. CD14+ TAMs isolated from mCRC inhibited T-cell proliferation, illustrating the immune suppressive phenotype of these cells. Conclusions: Inflammatory monocytes correlate to prognosis in mCRC. Therefore, targeting CCR2+ myeloid cells may improve anti-tumor immunity and patient survival in metastatic disease.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1697-1697
Author(s):  
Shiv K. Gupta ◽  
Mamta Gupta ◽  
Barbara Hoffman ◽  
Dan A. Liebermann

Abstract Growth arrest and DNA damage, Gadd45 gene family members are rapidly induced by genotoxic agents as well as by apoptosis and differentiation inducing cytokines. Their role in hemetopoiesis, wherein proliferation, differentiation and apoptosis integrate to maintain cellular homeostasis, is not clear. Using bone marrow cells from gadd45a or gadd45b deficient and wild type littermate mice we have investigated the role of Gadd45 proteins in cytokine induced myeloid cell differentiation in vitro. Bone marrow cells obtained from either gadd45a or gadd45b deficient mice displayed compromised cytokines (IL3, GM-CSF, M-CSF or G-CSF) induced myelopoiesis, resulting in a quantitatively decreased population of mature myeloid cells. Immuno-phenotyping with antibodies to cell surface molecules associated with myeloid cell maturation confirmed impaired myeloid cell maturation in Gadd45a or b deficient bone marrow cells treated with the above cytokines. Analysis of apoptosis by annexin-V and PI staining followed by FACS analysis showed a substantially higher apoptosis in Gadd45a−/− as well as gadd45b−/− cells compared to wild type cells after treatment with M-CSF or G-CSF. Gadd45a−/− as well as gadd45b−/− bone marrow cells were found to be less clonogenic in methylcellulose medium. Morphologically compact and round colonies consisting of immature myeloid cells prevailed over dispersed- colonies consisting of mature myeloid cells in gadd45- deficient cells cultured in methyl cellulose containing IL-3. Furthermore, colony re-plating assay showed better self-renewal abilities in gadd45a−/− as well as gadd45b−/− progenitors, compared to wild type progenitor cells. Altered myelopoiesis in gadd45 a or b deficient mice was further confirmed in vivo by intra-peritoneal administration of sodium casienate - a known inducer of inflammatory response and myelopoiesis in mice bone marrow. Sodium casienate failed to enhance myelopoiesis in gadd45a or gadd45b deficient mice bone marrow, while wild type littermate mice showed a rapid induction of myelopoiesis. Simultaneously peritoneal exudates collected from gadd45 deficient mice consisted of 2–3 fold less myeloid cells compared to age matched wild type control mice after sodium casienate treatment. Gadd45a−/− or gadd45b−/− mice showed a slow recovery after myelo-suppressive effect of antimetabolite 5-Fluorouracil, which further confirmed that gadd45 deficiency leads to delayed myelopoiesis in mouse. Mechanistic aspects of Gadd45 deficiency, which results in impaired myelopoiesis are under investigation.


Sign in / Sign up

Export Citation Format

Share Document