scholarly journals 20(S)-Protopanaxdiol Suppresses the Abnormal Granule-Monocyte Differentiation of Hematopoietic Stem Cells in 4T1 Breast Cancer-Bearing Mouse

2020 ◽  
Vol 2020 ◽  
pp. 1-11 ◽  
Author(s):  
Wen-Qin Guo ◽  
Ying-Ge Chen ◽  
Rong-Zhen Shi ◽  
Kai He ◽  
Jian-Feng Wang ◽  
...  

Panax notoginseng (PN) has been used as a qi- and blood-activating (Huoxue) drug for thousands of years in China. It has also been widely used as an anticancer drug at present. As a Huoxue drug, the effect of PN on hematopoietic differentiation in tumor-bearing body has been paid more and more attention. Our research found that panax notoginseng saponins (PNS), especially panaxadiol saponins (PDS) and its aglucon 20(S)-Protopanaxdiol (PPD), could improve the immunosuppressive state by regulating the abnormal hematopoietic differentiation in a tumor-bearing body by multiple ways. An interesting phenomenon is that PDS reduced the neutrophil-lymphocyte ratio (NLR) via its inhibition effect on the granule-monocyte differentiation of spleen cells, which is associated with a decrease in the secretion of tumor MPO, G-CSF, PU.1, and C/EBPα. Otherwise, PDS increased the proportion of both hematopoietic stem cells and erythroid progenitor cells in the bone marrow, but inhibited spleen erythroid differentiation via inhibiting secretion of tumor EPO, GATA-1, and GATA-2. This study suggests that PNS regulated the tumor-induced abnormal granule-monocyte differentiation of hematopoietic stem cells, affecting the distribution and function of haemocytes in tumor-bearing mice.

2020 ◽  
Author(s):  
Jicong Du ◽  
Penglin Xia ◽  
Yuan Gao ◽  
Ying Cheng ◽  
Ruling Liu ◽  
...  

Abstract Background: Hematopoiesis and the differentiation of HSC have been proved to not only play important roles in cancer progression but also be changed or reprogrammed by the tumor microenvironment itself. In this study, we investigated the changes of HSCs differentiation in advanced tumor-bearing mice. Methods: The tumor-bearing mice model was established by subcutaneously inoculating with xenografts of B16-F10 mouse melanoma cells into the right back of male wild-type C57BL/6 mice. Hematopoietic stem cells and multilineage differentiation were evaluated using blood routine, HE-staining, flow cytometry assay and HSCs culture techniques. Results: The multilineage differentiation of hematopoietic stem cells was reprogrammed in vivo . Especially, the differentiations of megakaryocyte and erythrocyte were blocked , while myeloid cell and lymphoid cell differentiation was encouraged in advanced tumor-bearing mice. Conclusion: In this study we showed the potential mechanism of hematopoietic disorder in tumor condition from a respective of hematopoietic stem cell and multilineage differentiation, which provided new knowledge regarding cachexia.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1486-1486
Author(s):  
Marie-Claude Gaudreau ◽  
Ehssan Sharif Askari ◽  
Florian Heyd ◽  
Tarik Moroy

Abstract Abstract 1486 Poster Board I-509 Hematopoietic differentiation has to be tightly regulated since uncontrolled or exaggerated development of blood cells may lead to the development of leukemia or autoimmune diseases. Many mechanisms exist to control hematopoiesis on a molecular level, including the regulation of transcription, which has been intensely studied. However, new evidence suggests the process of alternative splicing to be an important regulator of the maturation and activation of blood- and immune effector cells. One of the factors that has been identified as a potential regulator of the immune response and controls alternative splicing is “heterogenous nuclear ribonucleoprotein L” (hnRNP L). This factor affects among others the alternative splicing of the CD45 gene, which encodes the major tyrosine phosphatase expressed on all hematopoietic cells. To investigate the biological role of hnRNP L as a regulator of alternative splicing in hematopoiesis, we have generated conditional hnRNP L knockout (KO) mice carrying floxed alleles that can be deleted by co expression of Cre recombinase. Both the inducible MxCre transgene or Vav-Cre transgene, which is active in all hematopoietic cells were introduced into hnRNP Lfl/fl mice. We found that the conditional deletion of hnRNP L by the Vav Cre transgene led to early mortality before birth (at stage E17.5) and flow cytometric analysis of fetal liver of hnRNP Lfl/fl, Vav-Cre mice or bone marrow from pIpC induced hnRNP Lfl/fl Mx-Cre mice showed a deficit in erythrocyte maturation. In addition, fetal thymi from hnRNP Lfl/fl X Vav-Cre mice were severely reduced in cellularity and showed disturbed T cell maturation. Moreover, the deletion of hnRNP L results in reduced numbers of Lin−Sca1+ckit+ (LSK) cells, common lymphoid progenitors (CLPs), common myeloid progenitors (CMPs), granulocyte-monocyte progenitors (GMPs) and megakaryocyte-erythrocyte progenitors (MEPs). Strikingly, while most of the progenitors and the short-term hematopoietic stem cells (HSCs) were affected by the deletion of hnRNP L, the population of long term HSCs was not reduced. We found a high percentage of Annexin V positive cells in the LSK population suggesting that the loss of progenitors and short term HSCs in hnRNP L deficient mice is due to an accelerated cell death. To test whether stem cells lacking hnRNP L were still functional, we sorted Lin−Sca1+ckit+ (LSK) cells and cultured them on either methylcellulose or the feeder cell lines OP9 and OP9-DL1. The co-culture with OP9 or OP9-DL1 cells demonstrated that hnRNP L−/− LSK cells had lost their potential to differentiate into B and T lymphocytes. Similarly, hnRNP L deficient LSK cells were unable to give rise to lymphoid, myeloid or erythroid colonies on methylcellulose. This suggests that hnRNP L is required to maintain not only the numbers of hematopoietic stem cells, but also their ability for multilineage differentiation. We conclude that the regulation of alternative splicing is an essential component of the regulatory network required to maintain hematopoietic differentiation and the functional integrity of hematopoietic stem cells. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 382 (1) ◽  
pp. 111445
Author(s):  
Fabiana Mammoli ◽  
Sandra Parenti ◽  
Mariana Lomiento ◽  
Claudia Gemelli ◽  
Claudio Giacinto Atene ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (10) ◽  
pp. 2097-2106 ◽  
Author(s):  
Yinshi Guo ◽  
Chao Niu ◽  
Peter Breslin ◽  
Minghui Tang ◽  
Shubin Zhang ◽  
...  

Abstract It has been found that c-Myc protein plays a critical role in controlling self-renewal versus differentiation in hematopoietic stem cells. We report that c-Myc also controls the fate of megakaryocyte-erythrocyte progenitors through regulating the differentiation of erythroid and megakaryocytic progenitors. In addition to the significant reduction of granulocytes/macrophages and B and T lymphocytes because of the reduction of their corresponding progenitors, we found significantly increased numbers of megakaryocytic progenitors and mature megakaryocytes in bone marrow and spleens of c-Myc-knockout (c-Myc−/−) mice. Differentiation of erythrocytes was blocked at the erythroid progenitor stage. This increased megakaryocytopoiesis is a cell-intrinsic defect of c-Myc-mutant hematopoietic stem cells, as shown by transplantation studies. Furthermore, we found that c-Myc is required for polyploidy formation but not for cytoplasmic maturation of megakaryocytes. Megakaryocytes from c-Myc−/− mice are significantly smaller in size and lower in ploidy than those of control mice; however, because of the dramatic increase in megakaryocyte number, although fewer platelets are produced by each megakaryocyte, a greater than 3-fold increase in platelet number was consistently observed in c-Myc−/− mice. Thus, c-Myc−/− mice develop a syndrome of severe thrombocytosis-anemia-leukopenia because of significant increases in megakaryocytopoiesis and concomitant blockage of erythrocyte differentiation and reductions in myelolymphopoiesis.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2465-2465
Author(s):  
Ksenia Magidey ◽  
Ksenya Kveler ◽  
Rachelly Normand ◽  
Tongwu Zhang ◽  
Michael Timaner ◽  
...  

Metastasis is the major cause of death in cancer patients. Recent studies have demonstrated that the crosstalk between different host and tumor cells in the tumor microenvironment regulates tumor progression and metastasis. Specifically, immune cell myeloid skewing is a prominent promoter of metastasis. While previous studies have demonstrated that the recruitment of myeloid cells to tumors is a critical step in dictating tumor fate, the reservoir of these cells in the bone marrow (BM) compartment and their differentiation pattern has not been explored. Here we utilized a unique model system consisting of tumor cell clones with low and high metastatic potential (met-low and met-high, respectively) derived from melanoma and breast carcinoma cell lines. Hematopoietic stem cells (HSCs) and their early progenitor subset were defined as Lin-/Sca1+/CD117+, representing LSK cells. BM transplantation experiments using GFP-positive LSK cells derived from met-low and met-high tumor bearing mice were carried out to study lineage differentiation. The genetic signatures of LSK cells were analyzed by single cell RNA-sequencing (scRNA-seq). This analysis included unbiased automated annotation of individual cell types by correlating single-cell gene expression with reference transcriptomic data sets (SingleR algorithm) in order to evaluate the proportions of cell types in BM and reveal cell type-specific differentially expressed genes. Expression patterns of proteins originated from tumor cells were analyzed using a range of multi-omics techniques including nanostring, protein array, and mass spectrometry analysis. Tumor proteomic data was integrated with differential receptor expression patterns in BM cell types to reveal novel crosstalk between tumor cells and HSCs in the BM compartment. Mice bearing met-high tumors exhibited a significant increase in the percentage of LSK cells in the BM in comparison to tumor-free mice or mice bearing met-low tumors. These results were confirmed by functional CFU assays of peripheral blood of met-high compared to met-low tumor bearing mice. In addition, mice that underwent BM transplantation with GFP-positive LSK cells obtained from met-high inoculated donors exhibited an increased percentage of circulating GFP-positive myeloid cells in comparison to counterpart mice transplanted with LSK cells from met-low inoculated donors. Moreover, scRNA-seq analysis of LSK cells obtained from the BM of met-low and met-high tumor bearing mice revealed that met-high tumors induce the enrichment of monocyte-dendritic progenitor population (MDP), confirmed also by flow cytometry. To uncover the possible factors involved in myeloid programming of LSK cells, we performed a proteomic screen of tumor conditioned medium and integrated the results with the scRNA-seq data analysis. This analysis revealed that the IL-6-IL-6R axis is highly active in LSK-derived MDP cells from mice bearing met-high tumors. An adoptive transfer experiment using MDP-GFP+ cells obtained from BM of met-high tumor bearing mice demonstrated that met-high tumors directly dictate HSC fate decision towards myeloid bias, resulting in increased metastasis. Evidently, blocking IL-6 in mice bearing met-high tumors reduced the number of MDP cells, and consequently decreased metastasis. Our study reveals a unique crosstalk between tumor cells and HSCs. It provides new insight into the mechanism by which tumors contribute to the presence of supporting stroma. Specifically, tumors secreting IL-6 dictate a specific genetic signature in HSCs that programs them towards myeloid differentiation, thereby inducing a metastatic switch. Disclosures No relevant conflicts of interest to declare.


2018 ◽  
Vol 19 (8) ◽  
pp. 2353 ◽  
Author(s):  
Michel Aurrand-Lions ◽  
Stéphane Mancini

After birth, the development of hematopoietic cells occurs in the bone marrow. Hematopoietic differentiation is finely tuned by cell-intrinsic mechanisms and lineage-specific transcription factors. However, it is now clear that the bone marrow microenvironment plays an essential role in the maintenance of hematopoietic stem cells (HSC) and their differentiation into more mature lineages. Mesenchymal and endothelial cells contribute to a protective microenvironment called hematopoietic niches that secrete specific factors and establish a direct contact with developing hematopoietic cells. A number of recent studies have addressed in mouse models the specific molecular events that are involved in the cellular crosstalk between hematopoietic subsets and their niches. This has led to the concept that hematopoietic differentiation and commitment towards a given hematopoietic pathway is a dynamic process controlled at least partially by the bone marrow microenvironment. In this review, we discuss the evolving view of murine hematopoietic–stromal cell crosstalk that is involved in HSC maintenance and commitment towards B cell differentiation.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1649-1649 ◽  
Author(s):  
Ayana Kon ◽  
Satoshi Yamazaki ◽  
Yasunori Ota ◽  
Keisuke Kataoka ◽  
Yusuke Shiozawa ◽  
...  

Abstract Recent genetic studies have revealed frequent and specific pathway mutations involving multiple components of the RNA splicing machinery in myelodysplasia. Among these, SRSF2 mutations are more prevalent in CMML subtype and are associated with poor prognosis. Mutations showed a prominent hotspot involving proline 95, causing either P95H, P95L, or P95 conversion. Comprehensive analysis in our large cohort of MDS revealed that SRSF2 mutations showed a significant trend to coexist with TET2, STAG2, ASXL1 and RUNX1 mutations, while being mutually exclusive with EZH2 mutations. On the other hand, the molecular mechanism by which SRSF2 mutations lead to myelodysplasia remains largely unknown.  To elucidate the role of SRSF2 mutations in the development of myelodysplasia, we generated a heterozygous conditional knock-in mouse model of Srsf2 P95H mutation and crossed them with Vav1-Cre transgenic mice. Srsf2 P95H mutant mice exhibited macrocytic anemia, otherwise no significant changes in total peripheral blood (PB) cell counts compared to wild-type mice at 8-15 weeks after birth. There was no significant difference in lineage composition as well as blood cell morphology between wild-type and mutant mice in both bone marrow (BM) and PB. Flow cytometry of BM cells showed significant decrease of the number of hematopoietic stem cells (HSCs) and multipotent progenitor cells defined as Lin-Sca-1+Kit+ (LSK) fractions in Srsf2 P95H mice compared to wild-type mice. On the other hand, there were no significant differences in the number of more differentiated progenitor cells including common myeloid progenitors (CMPs), granulocyte/macrophage lineage-restricted progenitors (GMPs), megakaryocyte/erythrocyte lineage-restricted progenitors (MEPs), and common lymphoid progenitors (CLPs) between Srsf2 P95H and wild-type mice. These observations suggested that heterozygous Srsf2 mutation led to deregulation of hematopoietic stem cells, which however, is not sufficient for the development of MDS.  We next performed noncompetitive transplantation experiments to assess the cell intrinsic effects of Srsf2 P95H mutations. In PB, decreased white blood cell counts and progressive anemia were observed in mutant mice, which were evident as early as one month after transplantation. Cytological analysis of PB revealed morphological abnormalities in mice reconstituted with Srsf2 mutated cells, including hypersegmentation in neutrophils and dysplasia in the erythroid lineage. Srsf2 mutant-reconstituted mice showed normo-to-hypercellular marrow, where abnormal megakaryocyte distribution adjacent to trabecular bone and erythroid dysplasia was observed. Flow cytometrical analysis revealed decreased numbers of HSCs, LSK fractions and CMPs, whereas there was no significant change in the number of MEPs, GMPs and CLPs in BM. The BM erythroid progenitors were decreased in mutant-reconstituted mice, whereas the mutant mice showed splenic erythropoiesis with increased erythroid progenitors, suggesting the presence of extramedullary hematopoiesis, which was not seen in wild-type Srsf2 transduced mice. These observations suggested that the Srsf2 mutation led to ineffective hematopoiesis and morphological abnormalities, which seemed to recapitulate the phenotype of MDS.  Subsequently, we assessed the reconstitution capacity of whole BM cells from Srsf2 mutant mice in competitive transplantation experiments. The donor chimerism of Srsf2 P95H-derived cells in PB was significantly lower than that of wild-type cells. At 4 months post transplantation, the chimerism of Srsf2 P95H-derived cells was remarkably lower than that of wild-type cells in the fractions of HSCs, MPPs, CMPs, MEPs, GMPs and CLPs in BM. Furthermore, the reduced donor chimerism for Srsf2 P95H mutants was recapitulated in secondary transplantation experiments.  In summary, our results demonstrated that heterozygous P95H mutation of Srsf2 led to deregulation of hematopoietic stem cells that was evident from reduced competitive repopulation and impaired hematopoietic differentiation. Whereas mice reconstituted with Srsf2 mutant BM cells developed MDS-like phenotype in non-competitive transplantation setting, Srsf2 mutation by itself does not seem to be sufficient to develop MDS without transplantation, raising the possibility that an additional genetic and/or epigenetic events was required for overt MDS phenotype. Disclosures No relevant conflicts of interest to declare.


Hepatology ◽  
2000 ◽  
Vol 31 (6) ◽  
pp. 1251-1256 ◽  
Author(s):  
Lucy Golden-Mason ◽  
Michael P. Curry ◽  
Niamh Nolan ◽  
Oscar Traynor ◽  
Gerry McEntee ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document