72 results on '"Ewa Sitnicka"'
Search Results
2. Ontogenic shifts in cellular fate are linked to proteotype changes in lineage-biased hematopoietic progenitor cells
- Author
-
Maria Jassinskaja, Kristýna Pimková, Nejc Arh, Emil Johansson, Mina Davoudi, Carlos-Filipe Pereira, Ewa Sitnicka, and Jenny Hansson
- Subjects
fetal hematopoiesis ,adult hematopoiesis ,proteomics ,hematopoietic progenitor cells ,lymphopoiesis ,myelopoiesis ,Biology (General) ,QH301-705.5 - Abstract
Summary: The process of hematopoiesis is subject to substantial ontogenic remodeling that is accompanied by alterations in cellular fate during both development and disease. We combine state-of-the-art mass spectrometry with extensive functional assays to gain insight into ontogeny-specific proteomic mechanisms regulating hematopoiesis. Through deep coverage of the cellular proteome of fetal and adult lympho-myeloid multipotent progenitors (LMPPs), common lymphoid progenitors (CLPs), and granulocyte-monocyte progenitors (GMPs), we establish that features traditionally attributed to adult hematopoiesis are conserved across lymphoid and myeloid lineages, whereas generic fetal features are suppressed in GMPs. We reveal molecular and functional evidence for a diminished granulocyte differentiation capacity in fetal LMPPs and GMPs relative to their adult counterparts. Our data indicate an ontogeny-specific requirement of myosin activity for myelopoiesis in LMPPs. Finally, we uncover an ontogenic shift in the monocytic differentiation capacity of GMPs, partially driven by a differential expression of Irf8 during fetal and adult life.
- Published
- 2021
- Full Text
- View/download PDF
3. Editorial: Molecular and Cellular Pathways in NK Cell Development
- Author
-
Ewa Sitnicka, Yenan Bryceson, Aharon G. Freud, and Emily M. Mace
- Subjects
NK cells ,developmental and maturation stages ,regulatory pathways ,disease-induced defects ,clinical applications ,Immunologic diseases. Allergy ,RC581-607 - Published
- 2020
- Full Text
- View/download PDF
4. Term amniotic fluid: an unexploited reserve of mesenchymal stromal cells for reprogramming and potential cell therapy applications
- Author
-
Roksana Moraghebi, Agnete Kirkeby, Patricia Chaves, Roger E. Rönn, Ewa Sitnicka, Malin Parmar, Marcus Larsson, Andreas Herbst, and Niels-Bjarne Woods
- Subjects
Term amniotic fluid ,Caesarean section deliveries ,Mesenchymal stromal cells ,Cell-based therapy ,Cellular reprogramming ,Pluripotency ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Mesenchymal stromal cells (MSCs) are currently being evaluated in numerous pre-clinical and clinical cell-based therapy studies. Furthermore, there is an increasing interest in exploring alternative uses of these cells in disease modelling, pharmaceutical screening, and regenerative medicine by applying reprogramming technologies. However, the limited availability of MSCs from various sources restricts their use. Term amniotic fluid has been proposed as an alternative source of MSCs. Previously, only low volumes of term fluid and its cellular constituents have been collected, and current knowledge of the MSCs derived from this fluid is limited. In this study, we collected amniotic fluid at term using a novel collection system and evaluated amniotic fluid MSC content and their characteristics, including their feasibility to undergo cellular reprogramming. Methods Amniotic fluid was collected at term caesarean section deliveries using a closed catheter-based system. Following fluid processing, amniotic fluid was assessed for cellularity, MSC frequency, in-vitro proliferation, surface phenotype, differentiation, and gene expression characteristics. Cells were also reprogrammed to the pluripotent stem cell state and differentiated towards neural and haematopoietic lineages. Results The average volume of term amniotic fluid collected was approximately 0.4 litres per donor, containing an average of 7 million viable mononuclear cells per litre, and a CFU-F content of 15 per 100,000 MNCs. Expanded CFU-F cultures showed similar surface phenotype, differentiation potential, and gene expression characteristics to MSCs isolated from traditional sources, and showed extensive expansion potential and rapid doubling times. Given the high proliferation rates of these neonatal source cells, we assessed them in a reprogramming application, where the derived induced pluripotent stem cells showed multigerm layer lineage differentiation potential. Conclusions The potentially large donor base from caesarean section deliveries, the high yield of term amniotic fluid MSCs obtainable, the properties of the MSCs identified, and the suitability of the cells to be reprogrammed into the pluripotent state demonstrated these cells to be a promising and plentiful resource for further evaluation in bio-banking, cell therapy, disease modelling, and regenerative medicine applications.
- Published
- 2017
- Full Text
- View/download PDF
5. FLT3-ITDs Instruct a Myeloid Differentiation and Transformation Bias in Lymphomyeloid Multipotent Progenitors
- Author
-
Adam J. Mead, Shabnam Kharazi, Deborah Atkinson, Iain Macaulay, Christian Pecquet, Stephen Loughran, Michael Lutteropp, Petter Woll, Onima Chowdhury, Sidinh Luc, Natalija Buza-Vidas, Helen Ferry, Sally-Ann Clark, Nicolas Goardon, Paresh Vyas, Stefan N. Constantinescu, Ewa Sitnicka, Claus Nerlov, and Sten Eirik W. Jacobsen
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Whether signals mediated via growth factor receptors (GFRs) might influence lineage fate in multipotent progenitors (MPPs) is unclear. We explored this issue in a mouse knockin model of gain-of-function Flt3-ITD mutation because FLT3-ITDs are paradoxically restricted to acute myeloid leukemia even though Flt3 primarily promotes lymphoid development during normal hematopoiesis. When expressed in MPPs, Flt3-ITD collaborated with Runx1 mutation to induce high-penetrance aggressive leukemias that were exclusively of the myeloid phenotype. Flt3-ITDs preferentially expanded MPPs with reduced lymphoid and increased myeloid transcriptional priming while compromising early B and T lymphopoiesis. Flt3-ITD-induced myeloid lineage bias involved upregulation of the transcription factor Pu.1, which is a direct target gene of Stat3, an aberrantly activated target of Flt3-ITDs, further establishing how lineage bias can be inflicted on MPPs through aberrant GFR signaling. Collectively, these findings provide new insights into how oncogenic mutations might subvert the normal process of lineage commitment and dictate the phenotype of resulting malignancies.
- Published
- 2013
- Full Text
- View/download PDF
6. 3191 – DELINEATING HUMAN NATURAL KILLER CELL DEVELOPMENT THROUGH AN IN VITRO CELL- DIFFERENTIATION MAP AND BARCODE-CLONAL TRACING
- Author
-
Quinn, Ewa Sitnicka, primary, Vo, Dang Nghiem, additional, Yuan, Ouyang, additional, Dushime, Gladys Telliam, additional, Kotova, Olga, additional, and Bryder, David, additional
- Published
- 2022
- Full Text
- View/download PDF
7. 3204 – NK CELL-DEFECTS OCCURRING ALREADY AT THE PRE-MDS STAGE IN NUP98-HOXD13 TRANSGENIC MOUSE MODEL
- Author
-
Gladys Telliam Dushime, Ouyang Yuan, Olga Kotova, Maciej Ciesla, Henrik Lilljebjörn, Jonas Ungerbäck, Thoas Fioretos, Cristian Bellodi, David Bryder, and Ewa Sitnicka
- Subjects
Cancer Research ,Genetics ,Cell Biology ,Hematology ,Molecular Biology - Published
- 2022
- Full Text
- View/download PDF
8. Loss of MafA and MafB expression promotes islet inflammation
- Author
-
David Bryder, Isabella Artner, Rashmi B. Prasad, Luis Sarmiento, Malin Fex, Ewa Sitnicka, Monika Dudenhöffer-Pfeifer, Luis Rodrigo Cataldo, P. Chaves, Tania Singh, Lisbeth Hansen, Sara Bsharat, Corrado M. Cilio, Jesper K. Colberg, and Dan Holmberg
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Maf Transcription Factors, Large ,T cell ,MafB Transcription Factor ,Receptors, Antigen, T-Cell ,Antigen-Presenting Cells ,lcsh:Medicine ,Autoimmunity ,Article ,03 medical and health sciences ,Gene Knockout Techniques ,Islets of Langerhans ,Mice ,0302 clinical medicine ,Immune system ,Antigen ,medicine ,Animals ,lcsh:Science ,Regulation of gene expression ,Inflammation ,B-Lymphocytes ,Multidisciplinary ,Chemistry ,Pancreatic islets ,lcsh:R ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,MAFB ,Mutation ,lcsh:Q ,Gene expression ,Signal transduction ,030217 neurology & neurosurgery ,CD8 ,Signal Transduction - Abstract
Maf transcription factors are critical regulators of beta-cell function. We have previously shown that reduced MafA expression in human and mouse islets is associated with a pro-inflammatory gene signature. Here, we investigate if the loss of Maf transcription factors induced autoimmune processes in the pancreas. Transcriptomics analysis showed expression of pro-inflammatory as well as immune cell marker genes. However, clusters of CD4+ T and B220+ B cells were associated primarily with adult MafA−/−MafB+/−, but not MafA−/− islets. MafA expression was detected in the thymus, lymph nodes and bone marrow suggesting a novel role of MafA in regulating immune-cell function. Analysis of pancreatic lymph node cells showed activation of CD4+ T cells, but lack of CD8+ T cell activation which also coincided with an enrichment of naïve CD8+ T cells. Further analysis of T cell marker genes revealed a reduction of T cell receptor signaling gene expression in CD8, but not in CD4+ T cells, which was accompanied with a defect in early T cell receptor signaling in mutant CD8+ T cells. These results suggest that loss of MafA impairs both beta- and T cell function affecting the balance of peripheral immune responses against islet autoantigens, resulting in local inflammation in pancreatic islets.
- Published
- 2019
- Full Text
- View/download PDF
9. Loss of Canonical Notch Signaling Affects Multiple Steps in NK Cell Development in Mice
- Author
-
Sten Eirik W. Jacobsen, Lilian Wittmann, Hanane Boukarabila, Claudia Peitzsch, Alya Zriwil, P. Chaves, and Ewa Sitnicka
- Subjects
Cytotoxicity, Immunologic ,0301 basic medicine ,endocrine system ,Cellular differentiation ,T cell ,Immunology ,Cell ,Notch signaling pathway ,Biology ,Lymphocyte Activation ,Mice ,03 medical and health sciences ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Lymphopoiesis ,Progenitor cell ,Cells, Cultured ,Mice, Knockout ,Receptors, Notch ,Chimera ,Cell growth ,Cell Differentiation ,Lymphoid Progenitor Cells ,Cell biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Immunoglobulin J Recombination Signal Sequence-Binding Protein ,Signal Transduction - Abstract
Within the hematopoietic system, the Notch pathway is critical for promoting thymic T cell development and suppressing the B and myeloid lineage fates; however, its impact on NK lymphopoiesis is less understood. To study the role of Notch during NK cell development in vivo, we investigated different NK cell compartments and function in Rbp-Jkfl/flVav-Cretg/+ mice, in which Rbp-Jk, the major transcriptional effector of canonical Notch signaling, was specifically deleted in all hematopoietic cells. Peripheral conventional cytotoxic NK cells in Rbp-Jk–deleted mice were significantly reduced and had an activated phenotype. Furthermore, the pool of early NK cell progenitors in the bone marrow was decreased, whereas immature NK cells were increased, leading to a block in NK cell maturation. These changes were cell intrinsic as the hematopoietic chimeras generated after transplantation of Rbp-Jk–deficient bone marrow cells had the same NK cell phenotype as the Rbp-Jk–deleted donor mice, whereas the wild-type competitors did not. The expression of several crucial NK cell regulatory pathways was significantly altered after Rbp-Jk deletion. Together, these results demonstrate the involvement of canonical Notch signaling in regulation of multiple stages of NK cell development.
- Published
- 2018
- Full Text
- View/download PDF
10. Concurrent stem- and lineage-affiliated chromatin programs precede hematopoietic lineage restriction
- Author
-
Fatemeh, Safi, Parashar, Dhapola, Sarah, Warsi, Mikael, Sommarin, Eva, Erlandsson, Jonas, Ungerbäck, Rebecca, Warfvinge, Ewa, Sitnicka, David, Bryder, Charlotta, Böiers, Ram Krishna, Thakur, and Göran, Karlsson
- Subjects
Gene Expression Regulation ,Cell Differentiation ,Cell Lineage ,Hematopoietic Stem Cells ,Megakaryocytes ,Chromatin ,General Biochemistry, Genetics and Molecular Biology - Abstract
The emerging notion of hematopoietic stem and progenitor cells (HSPCs) as a low-primed cloud without sharply demarcated gene expression programs raises the question on how cellular-fate options emerge and at which stem-like stage lineage priming is initiated. Here, we investigate single-cell chromatin accessibility of Lineage
- Published
- 2022
- Full Text
- View/download PDF
11. Ontogenic shifts in cellular fate are linked to proteotype changes in lineage-biased hematopoietic progenitor cells
- Author
-
Emil Johansson, Kristýna Pimková, Mina Davoudi, Nejc Arh, Carlos Filipe Pereira, Maria Jassinskaja, Jenny Hansson, and Ewa Sitnicka
- Subjects
0301 basic medicine ,Proteomics ,Myeloid ,Proteome ,adult hematopoiesis ,Biology ,myelopoiesis ,General Biochemistry, Genetics and Molecular Biology ,Monocytes ,Immunophenotyping ,fetal hematopoiesis ,03 medical and health sciences ,0302 clinical medicine ,Fetus ,Myosin ,medicine ,Animals ,Humans ,Cell Lineage ,Lymphopoiesis ,Progenitor cell ,lcsh:QH301-705.5 ,Myeloid Progenitor Cells ,rho-Associated Kinases ,Cell Differentiation ,lymphopoiesis ,Lymphoid Progenitor Cells ,Hematopoietic Stem Cells ,Cell biology ,Mice, Inbred C57BL ,Haematopoiesis ,Adult Stem Cells ,Kinetics ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,lcsh:Biology (General) ,Interferon Regulatory Factors ,Myelopoiesis ,IRF8 ,030217 neurology & neurosurgery ,hematopoietic progenitor cells ,Granulocytes - Abstract
Summary: The process of hematopoiesis is subject to substantial ontogenic remodeling that is accompanied by alterations in cellular fate during both development and disease. We combine state-of-the-art mass spectrometry with extensive functional assays to gain insight into ontogeny-specific proteomic mechanisms regulating hematopoiesis. Through deep coverage of the cellular proteome of fetal and adult lympho-myeloid multipotent progenitors (LMPPs), common lymphoid progenitors (CLPs), and granulocyte-monocyte progenitors (GMPs), we establish that features traditionally attributed to adult hematopoiesis are conserved across lymphoid and myeloid lineages, whereas generic fetal features are suppressed in GMPs. We reveal molecular and functional evidence for a diminished granulocyte differentiation capacity in fetal LMPPs and GMPs relative to their adult counterparts. Our data indicate an ontogeny-specific requirement of myosin activity for myelopoiesis in LMPPs. Finally, we uncover an ontogenic shift in the monocytic differentiation capacity of GMPs, partially driven by a differential expression of Irf8 during fetal and adult life.
- Published
- 2020
12. Ontogenic Shifts in Cellular Fate Are Linked to Proteotype Changes in Mouse Hematopoietic Progenitor Cells
- Author
-
Emil Johansson, Ewa Sitnicka, Kristýna Pimková, Jenny Hansson, and Maria Jassinskaja
- Subjects
Blood cell ,Haematopoiesis ,Cell type ,medicine.anatomical_structure ,Myeloid ,medicine ,Progenitor cell ,IRF8 ,Biology ,Proteomics ,Granulopoiesis ,Cell biology - Abstract
The process of hematopoiesis is subject to extensive ontogenic remodeling that is accompanied by alterations in cellular fate both during normal development and upon malignant transformation. Although the functional differences between fetal and adult hematopoiesis are well established, the responsible molecular mechanisms have long remained largely unexplored at the proteomic level. Here, we have applied state-of-the-art mass spectrometry to gain deep coverage of the proteome of 100,000 fetal and adult lympho-myeloid multipotent progenitors (LMPPs), common lymphoid progenitors (CLPs) and granulocyte-monocyte progenitors (GMPs). Our analysis resulted in the identification and quantification of 4189 proteins, with over 200 proteins per cell type displaying differential expression between the fetus and the adult. The proteomic data demonstrate that features traditionally attributed to adult hematopoiesis are conserved across lymphoid and myeloid lineages, while generic fetal features are considerably more prominent in LMPPs and CLPs than in GMPs. Furthermore, we reveal molecular and functional evidence for a diminished granulocyte differentiation capacity in fetal LMPPs and GMPs relative to their adult counterparts, and show indications of a differential requirement of myosin activity for granulopoiesis in fetal and adult LMPPs. We have additionally identified the transcription factor Irf8 as significantly lower expressed in fetal relative to adult GMPs, and shown that its expression pattern correlates with an altered capacity for monocytic differentiation in the fetal cells. Collectively, our work represents a significant advancement in the understanding of the molecular programs that govern ontogenic differences in early hematopoiesis and mature blood cell production.Key pointsIn-depth proteomics links intrinsic molecular programs to functional output of fetal and adult lineage-biased hematopoietic progenitorsMyelopoiesis-associated molecular programs and myeloid differentiation capacity are subject to considerable ontogenic remodeling
- Published
- 2020
- Full Text
- View/download PDF
13. Concurrent stem- and lineage-affiliated chromatin programs precede hematopoietic lineage restriction
- Author
-
Ewa Sitnicka, David Bryder, Göran Karlsson, Fatemeh Safi, Ram Krishna Thakur, Sarah Warsi, Charlotta Böiers, Parashar Dhapola, and Eva Erlandsson
- Subjects
Haematopoiesis ,education.field_of_study ,Transcription (biology) ,Gene expression ,Population ,Priming (immunology) ,Progenitor cell ,Stem cell ,Biology ,education ,Chromatin ,Cell biology - Abstract
SUMMARYThe emerging notion of hematopoietic stem- and progenitor cells (HSPCs) as a low-primed cloud without sharply demarcated gene expression programs raises the question on how cellular fate options emerge, and at which stem-like stage lineage priming is initiated. Here we investigated single-cell chromatin accessibility of Lineage−, cKit+, Sca1+(LSK) HSPCs spanning the early differentiation landscape. Application of a signal-processing algorithm to detect transition points corresponding to massive alterations in accessibility of 571 transcription factor-motifs revealed a population of LSK FMS-like tyrosine kinase 3(Flt3)intCD9highcells that concurrently display stem-like and lineage-affiliated chromatin signatures pointing to a simultaneous gain of both Lympho-Myeloid and Megakaryocyte-Erythroid programs. Molecularly and functionally, these cells position between stem cells and committed progenitors, display multi-lineage capacity in vitro and in vivo, but lack self-renewal activity. This integrative molecular analysis resolves the heterogeneity of cells along hematopoietic differentiation and permits investigation of chromatin-mediated transition between multipotency and lineage restriction.
- Published
- 2020
- Full Text
- View/download PDF
14. Deletion of Nemo-like Kinase in T Cells Reduces Single-Positive CD8
- Author
-
Renée, Daams, Wondossen, Sime, Karin, Leandersson, Ewa, Sitnicka, and Ramin, Massoumi
- Subjects
Mice, Knockout ,Thymocytes ,Cell Survival ,Lymphoid Enhancer-Binding Factor 1 ,Cell Differentiation ,Histone Deacetylase 1 ,CD8-Positive T-Lymphocytes ,Protein Serine-Threonine Kinases ,Lymphocyte Activation ,Mice, Inbred C57BL ,Wnt Proteins ,Mice ,T-Lymphocyte Subsets ,Animals ,Signal Transduction - Abstract
The β-catenin/Wnt signaling pathway plays an important role in all stages of T cell development. Nemo-like kinase (NLK) is an evolutionary conserved serine/threonine kinase and a negative regulator of the Wnt signaling pathway. NLK can directly phosphorylate histone deacetylase 1 (HDAC1), as well as T cell factor/lymphoid enhancer-binding factor (TCF/LEF), causing subsequent repression of target gene transcription. By engineering mice lacking NLK in early stages of T cell development, we set out to characterize the role NLK plays in T cell development and found that deletion of NLK does not affect mouse health or lymphoid tissue development. Instead, these mice harbored a reduced number of single-positive (SP) CD8
- Published
- 2020
15. Molecular and Cellular Pathways in NK Cell Development
- Author
-
Emily M. Mace, Ewa Sitnicka, Yenan T. Bryceson, and Aharon G. Freud
- Subjects
Cell growth ,Cellular pathways ,Biology ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
16. Direct role of<scp>FLT</scp>3 in regulation of early lymphoid progenitors
- Author
-
Trine A. Kristiansen, Sten Eirik W. Jacobsen, Claus Nerlov, Joan Yuan, Alya Zriwil, Charlotta Böiers, Ewa Sitnicka, and Lilian Wittmann
- Subjects
0301 basic medicine ,medicine.medical_specialty ,lymphoid development ,Bone Marrow Cells ,Thymus Gland ,Biology ,Recombination-activating gene ,Mice ,03 medical and health sciences ,fluids and secretions ,0302 clinical medicine ,lymphoid progenitors ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Animals ,Lymphopoiesis ,Progenitor cell ,FLT3 ,B cell ,Progenitor ,Homeodomain Proteins ,Mice, Knockout ,Fetus ,Hematology ,Haematological Malignancy ,Cell Differentiation ,hemic and immune systems ,haematopoiesis ,Lymphoid Progenitor Cells ,conditional knock‐out mouse model ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,fms-Like Tyrosine Kinase 3 ,030220 oncology & carcinogenesis ,embryonic structures ,Cancer research ,Research Paper - Abstract
Summary Given that FLT3 expression is highly restricted on lymphoid progenitors, it is possible that the established role of FLT3 in the regulation of B and T lymphopoiesis reflects its high expression and role in regulation of lymphoid‐primed multipotent progenitors (LMPPs) or common lymphoid progenitors (CLPs). We generated a Flt3 conditional knock‐out (Flt3 fl/fl) mouse model to address the direct role of FLT3 in regulation of lymphoid‐restricted progenitors, subsequent to turning on Rag1 expression, as well as potentially ontogeny‐specific roles in B and T lymphopoiesis. Our studies establish a prominent and direct role of FLT3, independently of the established role of FLT3 in regulation of LMPPs and CLPs, in regulation of fetal as well as adult early B cell progenitors, and the early thymic progenitors (ETPs) in adult mice but not in the fetus. Our findings highlight the potential benefit of targeting poor prognosis acute B‐cell progenitor leukaemia and ETP leukaemia with recurrent FLT3 mutations using clinical FLT3 inhibitors.
- Published
- 2018
- Full Text
- View/download PDF
17. Cellular Barcoding Links B-1a B Cell Potential to a Fetal Hematopoietic Stem Cell State at the Single-Cell Level
- Author
-
David Bryder, Alya Zriwil, Trine A. Kristiansen, Ewa Sitnicka, Elin Jaensson Gyllenbäck, Joan Yuan, Tomas Björklund, Shamit Soneji, and Jeremy A. Daniel
- Subjects
0301 basic medicine ,Cell Plasticity ,Immunology ,Cell ,Mice, Transgenic ,Biology ,Immunophenotyping ,Mice ,03 medical and health sciences ,Single-cell analysis ,medicine ,Animals ,Immunology and Allergy ,Cell Self Renewal ,B cell ,B-Lymphocytes ,RNA-Binding Proteins ,Hematopoietic stem cell ,Cell Differentiation ,Hematopoietic Stem Cells ,Lymphocyte Subsets ,Clone Cells ,Hematopoiesis ,Cell biology ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Haematopoiesis ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Animals, Newborn ,Liver ,Female ,Ectopic expression ,Single-Cell Analysis ,Stem cell - Abstract
Hematopoietic stem cells (HSCs) undergo a functional switch in neonatal mice hallmarked by a decrease in self-renewing divisions and entry into quiescence. Here, we investigated whether the developmental attenuation of B-1a cell output is a consequence of a shift in stem cell state during ontogeny. Using cellular barcoding for in vivo single-cell fate analyses, we found that fetal liver definitive HSCs gave rise to both B-1a and B-2 cells. Whereas B-1a potential diminished in all HSCs with time, B-2 output was maintained. B-1a and B-2 plasticity could be reinitiated in a subset of adult HSCs by ectopic expression of the RNA binding protein LIN28B, a key regulator of fetal hematopoiesis, and this coincided with the clonal reversal to fetal-like elevated self-renewal and repopulation potential. These results anchor the attenuation of B-1a cell output to fetal HSC behavior and demonstrate that the developmental decline in regenerative potential represents a reversible HSC state.
- Published
- 2016
- Full Text
- View/download PDF
18. 3088 – DISSECTING THE ONTOGENIC REMODELING OF THE PROTEOMIC LANDSCAPE AND FUNCTIONALITY OF HEMATOPOIETIC PROGENITOR CELLS IN NORMAL DEVELOPMENT AND LEUKEMIA
- Author
-
Maria Jassinskaja, Ugarit Daher, Kristýna Pimková, Mohamed Eldeeb, David Bryder, Jenny Hansson, Emil Johansson, and Ewa Sitnicka
- Subjects
Cancer Research ,Cell type ,Oncogene ,Context (language use) ,Cell Biology ,Hematology ,Biology ,Cell fate determination ,Cell biology ,Haematopoiesis ,Proteome ,Genetics ,Progenitor cell ,Molecular Biology ,Transcription factor - Abstract
The process of hematopoiesis is subject to extensive ontogenic remodeling that is accompanied by alterations in cellular fate both during normal development and disease. Although the functional differences between fetal and adult hematopoiesis are well established, the responsible molecular mechanisms have long remained largely unexplored on the proteomic level. In our latest work, we characterized and compared the proteomic makeup of fetal and adult HSPCs (Jassinskaja et al., 2017, Cell Reports). Here, we have utilized state-of-the-art mass spectrometry to gain deep coverage of the proteome of 100,000 fetal and adult lymphoid-primed multipotent progenitors (LMPPs), common lymphoid progenitors (CLPs) and granulocyte-monocyte progenitors (GMPs). Our analysis resulted in the identification and quantification of over 4000 proteins, with 200-300 proteins per cell type displaying differential expression between the fetus and the adult. We uncovered ontogenic changes in expression of several transcription factors with critical roles in hematopoiesis. Together with other proteins involved in lineage commitment and differentiation, we show that protein expression in fetal and adult hematopoietic progenitor cells is predictive of differential lineage potential. Our subsequent functional analyses further emphasize that the proteomic differences affect cell fate both in the context of normal differentiation and leukemic hematopoiesis driven by the MLL-ENL fusion oncogene. Collectively, our work represents a significant advancement in the understanding of the molecular programs that govern ontogenic differences in hematopoiesis and provides a solid foundation for future investigation of which factors are responsible for the difference in susceptibility and outcome of different leukemias in infants and in adults.
- Published
- 2020
- Full Text
- View/download PDF
19. Term amniotic fluid: an unexploited reserve of mesenchymal stromal cells for reprogramming and potential cell therapy applications
- Author
-
Niels-Bjarne Woods, Andreas Herbst, P. Chaves, Agnete Kirkeby, Roksana Moraghebi, Roger Emanuel Rönn, Ewa Sitnicka, Malin Parmar, and Marcus Larsson
- Subjects
0301 basic medicine ,Amniotic fluid ,Cell- and Tissue-Based Therapy ,Mesenchymal stromal cells ,Medicine (miscellaneous) ,Cell Separation ,Regenerative medicine ,Cell therapy ,Osteogenesis ,Pregnancy ,Term amniotic fluid ,lcsh:QD415-436 ,Induced pluripotent stem cell ,Neurons ,lcsh:R5-920 ,Adipogenesis ,Cell Differentiation ,Cellular reprogramming ,Cell biology ,Haematopoiesis ,Caesarean section deliveries ,Molecular Medicine ,Female ,Stem cell ,lcsh:Medicine (General) ,Reprogramming ,Biobanking ,Pluripotent Stem Cells ,Pluripotency ,Biology ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Cell Line ,lcsh:Biochemistry ,03 medical and health sciences ,Cell Adhesion ,Humans ,Cell-based therapy ,Cell Lineage ,Cell Proliferation ,Research ,Mesenchymal stem cell ,Infant, Newborn ,Epithelial Cells ,Mesenchymal Stem Cells ,Cell Biology ,Fibroblasts ,Amniotic Fluid ,Hematopoiesis ,030104 developmental biology ,Immunology - Abstract
Background Mesenchymal stromal cells (MSCs) are currently being evaluated in numerous pre-clinical and clinical cell-based therapy studies. Furthermore, there is an increasing interest in exploring alternative uses of these cells in disease modelling, pharmaceutical screening, and regenerative medicine by applying reprogramming technologies. However, the limited availability of MSCs from various sources restricts their use. Term amniotic fluid has been proposed as an alternative source of MSCs. Previously, only low volumes of term fluid and its cellular constituents have been collected, and current knowledge of the MSCs derived from this fluid is limited. In this study, we collected amniotic fluid at term using a novel collection system and evaluated amniotic fluid MSC content and their characteristics, including their feasibility to undergo cellular reprogramming. Methods Amniotic fluid was collected at term caesarean section deliveries using a closed catheter-based system. Following fluid processing, amniotic fluid was assessed for cellularity, MSC frequency, in-vitro proliferation, surface phenotype, differentiation, and gene expression characteristics. Cells were also reprogrammed to the pluripotent stem cell state and differentiated towards neural and haematopoietic lineages. Results The average volume of term amniotic fluid collected was approximately 0.4 litres per donor, containing an average of 7 million viable mononuclear cells per litre, and a CFU-F content of 15 per 100,000 MNCs. Expanded CFU-F cultures showed similar surface phenotype, differentiation potential, and gene expression characteristics to MSCs isolated from traditional sources, and showed extensive expansion potential and rapid doubling times. Given the high proliferation rates of these neonatal source cells, we assessed them in a reprogramming application, where the derived induced pluripotent stem cells showed multigerm layer lineage differentiation potential. Conclusions The potentially large donor base from caesarean section deliveries, the high yield of term amniotic fluid MSCs obtainable, the properties of the MSCs identified, and the suitability of the cells to be reprogrammed into the pluripotent state demonstrated these cells to be a promising and plentiful resource for further evaluation in bio-banking, cell therapy, disease modelling, and regenerative medicine applications. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0582-6) contains supplementary material, which is available to authorized users.
- Published
- 2017
- Full Text
- View/download PDF
20. Functional Resolution of Long-Term-and Short-Term- Hematopoietic Stem Cells
- Author
-
Carl Storey, Stephen H. Bartelmez, and Ewa Sitnicka
- Subjects
Haematopoiesis ,medicine.anatomical_structure ,Cell division ,Cancer stem cell ,Cell culture ,medicine ,Hematopoietic stem cell ,Stem cell ,Biology ,Embryonic stem cell ,Molecular biology ,Neural stem cell - Abstract
Several studies have demonstrated that the hematopoietic stem cell (HSC) compartment consists of long-term repopulating (LTR) and short-term repopulating (STR) HSC. Here we describe an improved purification approach that identifies both LTR- and STR- HSC as being lineage negative, c-kit positive, with very low Hoechst 33342 retention (Lin-, c-kit+, Holow). However, further selection of cells based on their differential retention of Rhodamine 123 resolves cells into LTR-HSC and STR-HSC. We show that our sort method highly enriches for LTR-HSC (Rhlow) and STR-HSC (Rhhigh), and demonstrate that the Rhlow cells as single transplanted cells are able to engraft 70% of mice in a competitive long-term repopulating assay. We then describe several in vitro assays that resolve Rhlow and Rhhigh cells based on the ability of single cells to survive, form clones, vary the time to their first cell division, express a high proliferative potential (HPP) or to generate HPP daughter cells at the 2- to 8-cell stage. In the presence of IL-3 alone, single Rhlow cells divided rarely and then formed only small clones (~8 cells). In contrast, Rhhigh readily divided in IL-3 alone and went on to form large clones (~10,000 cells). However, in the presence of IL-3+IL-6+SCF, both cell populations cloned in vitro with high efficiency (>90%), although the proportion of HPP clones was significantly higher in the Rhlow cell fraction (~90% vs ~40%). Furthermore, In addition, we show that the time required by Rhlow cells to undergo their first cell division in vitro is relatively non-synchronous and longer than that of Rhhigh cells. In addition, an analysis of daughter cells generated during the initial cell divsions of Rhlow or Rhhigh cells in vitro showed that expansion or maintenance of total HPP daughter cells occurred only in the Rhlow cell fraction. We measured the proliferative potential of daughter cells derived from single Rhlow and Rhhigh cells at the 2-8 cell stage. At the 2-cell stage, Rhlow cells generated an increased number of HPP daughter cells (↑1.4-fold) compared to Rhhigh cells that appeared to maintain the total number of HPP daughter cells (1.0-fold). However, by the 8-cell stage, the total number of HPP daughter cells generated by Rhlow cells expanded to nearly double that of starting HPP numbers (↑1.9 fold), compared to a decline in total HPP daughter cells in 8-cell Rhhigh clones (↓0.5 fold). Our studies at the 2-cell stage directly demonstrate symmetrical divisions (2 HPP per 2 daughter cells) that result in HPP expansion. Thus, these studies of growth factor responsivness of purified HSC (survival, cloning efficiency, time to the first cell division) and differentiation pathways (their ability to generate HPP daughter cells) identify means to differentiate LTR- and STR HSC in vitro.
- Published
- 2016
- Full Text
- View/download PDF
21. Lymphomyeloid Contribution of an Immune-Restricted Progenitor Emerging Prior to Definitive Hematopoietic Stem Cells
- Author
-
Christina Jensen, Marella F. T. R. de Bruijn, Ewa Sitnicka, Adam J. Mead, Elisa Gomez Perdiguero, Shabnam Kharazi, Qiaolin Deng, Deborah Atkinson, Frederic Geissmann, Luca Melchiori, Anne Hultquist, Iain C. Macaulay, Emanuele Azzoni, Joana Carrelha, Sidinh Luc, Annica Pontén, Joanna C. A. Green, Sten Eirik W. Jacobsen, Isabelle Godin, Tiphaine Bouriez-Jones, Tiago C. Luis, Petter S. Woll, Michael Lutteropp, Rickard Sandberg, Gemma Swiers, Charlotta Böiers, Hématopoïèse normale et pathologique (U1170 Inserm), Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), Böiers, C, Carrelha, J, Lutteropp, M, Luc, S, Green, J, Azzoni, E, Woll, P, Mead, A, Hultquist, A, Swiers, G, Perdiguero, E, Macaulay, I, Melchiori, L, Luis, T, Kharazi, S, Bouriez-Jones, T, Deng, Q, Pontén, A, Atkinson, D, Jensen, C, Sitnicka, E, Geissmann, F, Godin, I, Sandberg, R, de Bruijn, M, and Jacobsen, S
- Subjects
Male ,Myeloid ,Biology ,Polymerase Chain Reaction ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Animals ,Myeloid Cells ,Lymphocytes ,Lymphopoiesis ,Myeloid Cell ,Cells, Cultured ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,Interleukin 3 ,0303 health sciences ,Innate immune system ,Animal ,Gene Expression Regulation, Developmental ,Hematopoietic Stem Cell ,Cell Differentiation ,Cell Biology ,Hematopoietic Stem Cells ,Embryonic stem cell ,Endothelial stem cell ,Haematopoiesis ,[SDV.BDD.EO]Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,Molecular Medicine ,Lymphocyte ,Female ,Stem cell - Abstract
In jawed vertebrates, development of an adaptive immune-system is essential for protection of the born organism against otherwise life-threatening pathogens. Myeloid cells of the innate immune system are formed early in development, whereas lymphopoiesis has been suggested to initiate much later, following emergence of definitive hematopoietic stem cells (HSCs). Herein, we demonstrate that the embryonic lymphoid commitment process initiates earlier than previously appreciated, prior to emergence of definitive HSCs, through establishment of a previously unrecognized entirely immune-restricted and lymphoid-primed progenitor. Notably, this immune-restricted progenitor appears to first emerge in the yolk sac and contributes physiologically to the establishment of lymphoid and some myeloid components of the immune-system, establishing the lymphomyeloid lineage restriction process as an early and physiologically important lineage-commitment step in mammalian hematopoiesis. Copyright © 2013 Elsevier Inc. All rights reserved.
- Published
- 2013
- Full Text
- View/download PDF
22. Macrophage colony-stimulating factor receptor marks and regulates a fetal myeloid-primed B-cell progenitor in mice
- Author
-
Joanna C. A. Green, Charlotta Böiers, Alya Zriwil, Petter S. Woll, Lilian Wittmann, Sten Eirik W. Jacobsen, and Ewa Sitnicka
- Subjects
0301 basic medicine ,Macrophage colony-stimulating factor ,Myeloid ,Hematopoiesis and Stem Cells ,Immunology ,Biology ,Biochemistry ,03 medical and health sciences ,Mice ,Fetus ,hemic and lymphatic diseases ,medicine ,Animals ,Cell Lineage ,Progenitor cell ,B cell ,Progenitor ,Mice, Knockout ,Lymphopoiesis ,Precursor Cells, B-Lymphoid ,Cell Biology ,Hematology ,Embryonic stem cell ,Cell biology ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,embryonic structures ,Bone marrow - Abstract
Although it is well established that unique B-cell lineages develop through distinct regulatory mechanisms during embryonic development, much less is understood about the differences between embryonic and adult B-cell progenitor cells, likely to underpin the genetics and biology of infant and childhood PreB acute lymphoblastic leukemia (PreB-ALL), initiated by distinct leukemia-initiating translocations during embryonic development. Herein, we establish that a distinct subset of the earliest CD19(+) B-cell progenitors emerging in the E13.5 mouse fetal liver express the colony-stimulating factor-1 receptor (CSF1R), previously thought to be expressed, and play a lineage-restricted role in development of myeloid lineages, and macrophages in particular. These early embryonic CSF1R(+)CD19(+) ProB cells also express multiple other myeloid genes and, in line with this, possess residual myeloid as well as B-cell, but not T-cell lineage potential. Notably, these CSF1R(+) myeloid-primed ProB cells are uniquely present in a narrow window of embryonic fetal liver hematopoiesis and do not persist in adult bone marrow. Moreover, analysis of CSF1R-deficient mice establishes a distinct role of CSF1R in fetal B-lymphopoiesis. CSF1R(+) myeloid-primed embryonic ProB cells are relevant for infant and childhood PreB-ALLs, which frequently have a bi-phenotypic B-myeloid phenotype, and in which CSF1R-rearrangements have recently been reported.
- Published
- 2016
23. Identification of an NK/T cell–restricted progenitor in adult bone marrow contributing to bone marrow– and thymic-dependent NK cells
- Author
-
Yanjuan Tang, Hojjatollah Nozad Charoudeh, Corrado M. Cilio, Sten Eirik W. Jacobsen, Min Cheng, and Ewa Sitnicka
- Subjects
Myeloid ,T-Lymphocytes ,T cell ,Immunology ,Bone Marrow Cells ,Cell Separation ,Thymus Gland ,Biology ,Biochemistry ,Natural killer cell ,Mice ,Interleukin 21 ,medicine ,Animals ,Cell Lineage ,Progenitor cell ,Reverse Transcriptase Polymerase Chain Reaction ,Multipotent Stem Cells ,Cell Differentiation ,Cell Biology ,Hematology ,Flow Cytometry ,Hematopoietic Stem Cells ,Natural killer T cell ,Hematopoiesis ,Killer Cells, Natural ,Mice, Inbred C57BL ,Haematopoiesis ,medicine.anatomical_structure ,Bone marrow - Abstract
Although bone marrow (BM) is the main site of natural killer (NK)–cell development in adult mice, recent studies have identified a distinct thymic-dependent NK pathway, implicating a possible close link between NK- and T-cell development in adult hematopoiesis. To investigate whether a potential NK-/T-lineage restriction of multipotent progenitors might take place already in the BM, we tested the full lineage potentials of NK-cell progenitors in adult BM. Notably, although Lin−CD122+NK1.1−DX5− NK-cell progenitors failed to commit to the B and myeloid lineages, they sustained a combined NK- and T-cell potential in vivo and in vitro at the single-cell level. Whereas T-cell development from NK/T progenitors is Notch-dependent, their contribution to thymic and BM NK cells remains Notch-independent. These findings demonstrate the existence of bipotent NK-/T-cell progenitors in adult BM.
- Published
- 2010
- Full Text
- View/download PDF
24. Junctional Adhesion Molecule 2 Intensifies T Lymphopoiesis of Hematopoietic Stem Cells By Facilitating Notch/Delta Signaling
- Author
-
Shin Kaneko, Alya Zriwil, Valgardur Sigurdsson, Kenichi Miharada, Visnja Radulovic, Mark van der Garde, Ewa Sitnicka Quinn, and Svetlana Soboleva
- Subjects
Chemistry ,T cell ,Immunology ,Notch signaling pathway ,Cell Biology ,Hematology ,T lymphocyte ,Cell cycle ,Biochemistry ,Cell biology ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,Lymphopoiesis ,Stem cell - Abstract
Phenotypically described hematopoietic stem cells (HSCs) represent a functionally heterogeneous pool of primitive cells with conceivable potential to replenish and maintain the whole hematopoietic system. The diverse lineage potential of HSCs is supposed to play a significant role in the response to different kinds of hematopoietic stress. Since subcategorization of HSCs biased towards specific lineage(s) highly relies on the retrospective information, e.g. transplantation assay, exploring additional markers will allow us to understand further molecular mechanisms of HSC regulation such as activation and lineage choice but also the degree of correlation between them. Here, we show that the cell surface protein Junctional adhesion molecule 2 (Jam2) serves as an amplifier of the Notch/Delta signal thereby representing the higher T cell potential of HSCs. Flow cytometry analyses revealed that a subset of CD150+CD48-KSL cells in mouse bone marrow (BM) were positive for Jam2 (Jam2+HSC, 36.6 ±13.0%), while other Jam family member, Jam1 (F11r), was expressed on all HSCs and Jam3 was not detected. Transplantation assay using 30 Jam2+ or Jam2-HSCs revealed that Jam2+HSCs reconstituted lethally irradiated mice more efficiently than Jam2-HSCs (77.5 ±15.9 and 51.7 ±29.3% in peripheral blood, respectively). Lineage analyses revealed that Jam2+HSCs have a greater potential in lymphoid cell reconstitution, particularly T cells, whereas the chimerism in myeloid cells was not significantly different from Jam2-HSCs. This tendency of higher contribution to the T cell development was even more pronounced in the secondary transplantation experiments, where the contribution of Jam2+HSCs in T cells was close to 100%. Of note, most of Jam2+HSCs were in a dormant state, suggesting that the T cell potential of Jam2+HSCs is independent of the cell cycle progression. Jam2 has been reported to mediate the Notch signaling through an interaction with Jam1 (Kobayashi et al., Nature, 2014). In addition, Jam2+HSCs express Notch1 at a higher level than Jam2-HSCs (23.6 ±6.7 and 9.05 ±5.8%, respectively). We therefore analyzed the functional role of Jam2 in the Notch/Delta-oriented T cell production using a competitive feeder-free T cell culture system. At a low concentration of DLL1, that is insufficient to promote T cell production by itself, Jam2+HSCs effectively produced T cell lineages only in the presence of recombinant Jam1 protein, but not Jam2 or Jam3. In contrast, Jam2+HSCs did not require Jam1 protein with a higher concentration of DLL1. These differences were not observed with Jam2-HSCs, indicating that Jam2/Jam1 interaction amplifies Notch signal transduction and is crucial for the subsequent T cell specification of Jam2+HSCs. To elucidate the molecular signature of Jam2+HSCs, gene expression profiling was performed using a microarray analysis. Gene set enrichment analysis (GSEA) observed that Jam2+HSCs were significantly enriched for common lymphoid progenitor (CLP) and early T cell gene expression. Of note, Jam2+HSCs were also enriched for E2F target genes, G2M checkpoint genes and glycolysis related genes, which potentially explains the reason why Jam2+HSCs display a bivalent phenotype: being more dormant compared to Jam2-HSCs at the steady state but at the same time having the capacity to reconstitute more actively upon engraftment. Since Jam2 positivity correlates to T cell potential, we asked if altered T lymphopoietic environment affects the proportion of Jam2+HSCs. In vivo T cell depletion resulted in significantly higher frequency of Jam2+HSCs but not upon other stress inducers, such as 5-FU treatment, suggesting that the increase in Jam2+HSC pool was specifically due to the T cell deficiency. These findings indicate that the lack of T cells, which also means a requirement for immediate T cell replenishment, leads to an increase of Jam2+HSC fraction. Our findings suggest that Jam2 is the key protein that controls T lymphopoiesis by enhancing the Notch/Delta signal transduction via interaction with Jam1. It also means that the lineage balance particularly towards T lymphopoiesis might be regulated at a higher stage of hematopoietic hierarchy than currently understood. Thus, Jam2 is a new marker representing the T lymphocyte potential of HSCs, as the frequency of Jam2+HSCs sensitively reflects the state of the T cell environment. Disclosures No relevant conflicts of interest to declare.
- Published
- 2017
- Full Text
- View/download PDF
25. Distinct and Overlapping Patterns of Cytokine Regulation of Thymic and Bone Marrow-Derived NK Cell Development
- Author
-
Tadepally Lakshmikanth, Ewa Sitnicka, Petter Höglund, Yanjuan Tang, Hojjatollah Nozad Charoudeh, Min Cheng, Sten Eirik W. Jacobsen, and Petter Brodin
- Subjects
medicine.medical_treatment ,Immunology ,Cell ,Bone Marrow Cells ,Thymus Gland ,Biology ,Mice ,Interleukin 21 ,Lymphopenia ,medicine ,Animals ,Immunology and Allergy ,Receptor ,Interleukin-15 ,Mice, Knockout ,Cell growth ,Interleukin-7 ,Membrane Proteins ,Cell Differentiation ,Lymphoid Progenitor Cells ,Cell biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cytokine ,fms-Like Tyrosine Kinase 3 ,Bone marrow ,Cytokine receptor ,Homeostasis ,Interleukin Receptor Common gamma Subunit ,Signal Transduction - Abstract
Although bone marrow (BM) represents the main site for postnatal NK cell development, recently a distinct thymic-dependent NK cell pathway was identified. These studies were designed to investigate the role of cytokines in regulation of thymic NK cells and to compare with established regulatory pathways of BM-dependent NK cell compartment. The common cytokine receptor γ-chain (Il2rg) essential for IL-15-induced signaling, and FMS-like tyrosine kinase 3 (FLT3) receptor ligand (Flt3l) were previously identified as important regulatory pathways of the BM NK cell compartment based on lack of function studies in mice, however their complementary action remains unknown. By investigating mice double-deficient in Il2rg and Flt3l (Flt3l−/− Il2rg−/−), we demonstrate that FLT3L is important for IL2Rg-independent maintenance of both immature BM as well as peripheral NK cells. In contrast to IL-7, which is dispensable for BM but important for thymic NK cells, IL-15 has a direct and important role in both thymic and BM NK cell compartments. Although thymic NK cells were not affected in Flt3l−/− mice, Flt3l−/−Il2rg−/− mice lacked detectable thymic NK cells, suggesting that FLT3L is also important for IL-2Rg-independent maintenance of thymic NK cells. Thus, IL-2Rg cytokines and FLT3L play complementary roles and are indispensable for homeostasis of both BM and thymic dependent NK cell development, suggesting that the cytokine pathways crucial for these two distinct NK cell pathways are largely overlapping.
- Published
- 2009
- Full Text
- View/download PDF
26. From the Bone Marrow to the Thymus: The Road Map of Early Stages of T-Cell Development
- Author
-
Ewa Sitnicka
- Subjects
Polymers and Plastics ,T-Lymphocytes ,T cell ,Cellular differentiation ,Bone Marrow Cells ,Cell Differentiation ,Thymus Gland ,Biology ,medicine.disease ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,Bone Marrow ,Immunology ,medicine ,Animals ,Humans ,Bone marrow ,Lymphopoiesis ,Progenitor cell ,Stem cell ,General Environmental Science - Abstract
The thymus produces new T cells throughout life but has no self-renewing ability and requires replenishment and recruitment of progenitors derived from the bone marrow. Despite the progress in delineation of mature blood cell development several questions remain regarding T lymphopoiesis. Understanding the developmental stages from multipotent hematopoietic stem cells (HSCs) to the T-cell lineage-restricted progenitors has many potential clinical implications as it is important for understanding malignant transformation in T-cell cancer, accelerating T-cell regeneration after bone marrow transplantation and chemotherapy, and establishing new therapies to treat T-cell immune deficiencies. This review focuses on the steps leading from the HSCs in the bone marrow to the lineage committed T cells inside the thymus.
- Published
- 2009
- Full Text
- View/download PDF
27. Permissive roles of hematopoietin and cytokine tyrosine kinase receptors in early T-cell development
- Author
-
Tobias Rydén, Ewa Sitnicka, Sten Eirik W. Jacobsen, Anna Lübking, Charlotta Böiers, Shabnam Kharazi, Christina Jensen, and Mikael Sigvardsson
- Subjects
Thymic stromal lymphopoietin ,T-Lymphocytes ,Immunology ,Biochemistry ,Receptor tyrosine kinase ,Mice ,Thymic Stromal Lymphopoietin ,Animals ,Lymphopoiesis ,Receptors, Cytokine ,Mice, Knockout ,Receptors, Interleukin-7 ,biology ,Janus kinase 1 ,Interleukin-7 ,Membrane Proteins ,Receptor Protein-Tyrosine Kinases ,Cell Biology ,Hematology ,Hematopoiesis ,Tyrosine kinase 2 ,ROR1 ,biology.protein ,Cancer research ,Cytokines ,Stromal Cells ,Platelet-derived growth factor receptor ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Although several cytokines have been demonstrated to be critical regulators of development of multiple blood cell lineages, it remains disputed to what degree they act through instructive or permissive mechanisms. Signaling through the FMS-like tyrosine kinase 3 (FLT3) receptor and the hematopoietin IL-7 receptor alpha (IL-7Ralpha) has been demonstrated to be of critical importance for sustained thymopoiesis. Signaling triggered by IL-7 and thymic stromal lymphopoietin (TSLP) is dependent on IL-7Ralpha, and both ligands have been implicated in T-cell development. However, we demonstrate that, whereas thymopoiesis is abolished in adult mice doubly deficient in IL-7 and FLT3 ligand (FLT3L), TSLP does not play a key role in IL-7-independent or FLT3L-independent T lymphopoiesis. Furthermore, whereas previous studies implicated that the role of other cytokine tyrosine kinase receptors in T lymphopoiesis might not involve permissive actions, we demonstrate that ectopic expression of BCL2 is sufficient not only to partially correct the T-cell phenotype of Flt3l(-/-) mice but also to rescue the virtually complete loss of all discernable stages of early T lymphopoiesis in Flt3l(-/-)Il7r(-/-) mice. These findings implicate a permissive role of cytokine receptors of the hematopoietin and tyrosine kinase families in early T lymphopoiesis.
- Published
- 2008
- Full Text
- View/download PDF
28. Resolving the fetal to adult switch in B lymphopoiesis at the single cell level
- Author
-
Yuan, Joan, primary, Kristiansen, Trine, additional, Vanhee, Stijn, additional, Jaensson-Gyllenbäck, Elin, additional, Zriwil, Alya, additional, Doyle, Alexander, additional, Quinn, Ewa Sitnicka, additional, Lang, Stefan, additional, Soneji, Shamit, additional, and Bryder, David, additional
- Published
- 2017
- Full Text
- View/download PDF
29. Involvement of CCR9 at multiple stages of adult T lymphopoiesis
- Author
-
Min Cheng, William W. Agace, Marcus Svensson, William E. Jenkinson, Graham Anderson, Corrado M. Cilio, Jan Marsal, Heli Uronen-Hansson, Sten Eirik W. Jacobsen, and Ewa Sitnicka
- Subjects
medicine.medical_specialty ,Stromal cell ,T-Lymphocytes ,T cell ,Immunology ,Bone Marrow Cells ,Thymus Gland ,Biology ,Mice ,Receptors, CCR ,Internal medicine ,medicine ,Animals ,Immunology and Allergy ,Lymphopoiesis ,Progenitor cell ,Cells, Cultured ,Mice, Knockout ,Cell Differentiation ,Cell Biology ,Hematopoietic Stem Cells ,Cell biology ,Mice, Inbred C57BL ,Haematopoiesis ,Thymocyte ,medicine.anatomical_structure ,Endocrinology ,Stem cell ,CCL25 - Abstract
The chemokine CCL25 is constitutively expressed in the thymus, and its receptor CCR9 is expressed on subsets of developing thymocytes. Nevertheless, the function of CCL25/CCR9 in adult thymopoiesis remains unclear. Here, we demonstrate that purified CCR9−/− hematopoietic stem cells are deficient in their ability to generate all major thymocyte subsets including double-negative 1 (DN1) cells in competitive transfers. CCR9−/− bone marrow contained normal numbers of lineage− Sca-1+c-kit+, common lymphoid progenitors, and lymphoid-primed multipotent progenitors (LMPP), and CCR9−/− LMPP showed similar T cell potential as their wild-type (WT) counterparts when cultured on OP9–δ-like 1 stromal cells. In contrast, early thymic progenitor and DN2 thymocyte numbers were reduced in the thymus of adult CCR9−/− mice. In fetal thymic organ cultures (FTOC), CCR9−/− DN1 cells were as efficient as WT DN1 cells in generating double-positive (DP) thymocytes; however, under competitive FTOC, CCR9−/− DP cell numbers were reduced significantly. Similarly, following intrathymic injection into sublethally irradiated recipients, CCR9−/− DN cells were out-competed by WT DN cells in generating DP thymocytes. Finally, in competitive reaggregation thymic organ cultures, CCR9−/− preselection DP thymocytes were disadvantaged significantly in their ability to generate CD4 single-positive (SP) thymocytes, a finding that correlated with a reduced ability to form TCR-MHC-dependent conjugates with thymic epithelial cells. Together, these results highlight a role for CCR9 at several stages of adult thymopoiesis: in hematopoietic progenitor seeding of the thymus, in the DN-DP thymocyte transition, and in the generation of CD4 SP thymocytes.
- Published
- 2007
- Full Text
- View/download PDF
30. Crucial role of FLT3 ligand in immune reconstitution after bone marrow transplantation and high-dose chemotherapy
- Author
-
Sara Duarte, Sten Eirik W. Jacobsen, Hojjatollah Nozad, Min Cheng, Ewa Sitnicka, and Natalija Buza-Vidas
- Subjects
T-Lymphocytes ,Immunology ,Delayed Graft Function ,Antineoplastic Agents ,Biochemistry ,Mice ,Immune system ,medicine ,Animals ,Regeneration ,Lymphopoiesis ,Progenitor cell ,Bone Marrow Transplantation ,Mice, Knockout ,B-Lymphocytes ,Fetal Stem Cells ,business.industry ,Membrane Proteins ,Cell Biology ,Hematology ,Marginal zone ,Bone marrow purging ,Transplantation ,medicine.anatomical_structure ,Immune System ,Bone marrow ,Stem cell ,business - Abstract
Almost 5 decades after the first clinical transplantations, delayed immune reconstitution remains a considerable hurdle in bone marrow transplantation, and the mechanisms regulating immune reconstitution after transplantation remain to be established. Whereas adult fms-like tyrosine kinase 3 ligand–deficient (FL−/−) mice have reduced numbers of early B- and T-cell progenitors, they sustain close to normal levels of mature B and T cells. Herein, we demonstrate that adult bone marrow cells fail to reconstitute B-cell progenitors and conventional B cells in lethally irradiated FL−/− recipients, which also display delayed kinetics of T-cell reconstitution. Similarly, FL is essential for B-cell regeneration after chemotherapy-induced myeloablation. In contrast, fetal progenitors reconstitute B lymphopoiesis in FL−/− mice, albeit at reduced levels. A critical role of FL in adult B lymphopoiesis is further substantiated by an age-progressive decline in peripheral conventional B cells in FL−/− mice, whereas fetally and early postnatally derived B1 and marginal zone B cells are sustained in a FL-independent manner. Thus, FL plays a crucial role in sustaining conventional B lymphopoiesis in adult mice and, as a consequence, our findings implicate a critical role of FL in promoting immune reconstitution after myeloablation and bone marrow transplantation.
- Published
- 2007
- Full Text
- View/download PDF
31. Junctional Adhesion Molecule 2 Represents a Novel Subset of Hematopoietic Stem Cells Poised for T Lymphopoiesis
- Author
-
Radulovic, Visnja, primary, van der Garde, Mark, additional, Sigurdsson, Valgardur, additional, Zriwil, Alya, additional, Soboleva, Svetlana, additional, Kaneko, Shin, additional, Quinn, Ewa Sitnicka, additional, and Miharada, Kenichi, additional
- Published
- 2016
- Full Text
- View/download PDF
32. Identification of Flt3+ Lympho-Myeloid Stem Cells Lacking Erythro-Megakaryocytic Potential
- Author
-
Yutaka Sasaki, Mikael Sigvardsson, Anne Hultquist, David Bryder, Sten Eirik W. Jacobsen, Karina Liuba, Kristina Anderson, Jörgen Adolfsson, Robert Månsson, Liping Yang, O J Borge, Ewa Sitnicka, Natalija Buza-Vidas, Lina Thorén, and Christina Jensen
- Subjects
Lineage (genetic) ,Myeloid ,Biochemistry, Genetics and Molecular Biology(all) ,T cell ,Cellular differentiation ,Hematopoietic stem cell ,hemic and immune systems ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,medicine.anatomical_structure ,Myeloid stem cell ,hemic and lymphatic diseases ,Fms-Like Tyrosine Kinase 3 ,Immunology ,medicine ,Lymphoid Progenitor Cells - Abstract
All blood cell lineages derive from a common hematopoietic stem cell (HSC). The current model implicates that the first lineage commitment step of adult pluripotent HSCs results in a strict separation into common lymphoid and common myeloid precursors. We present evidence for a population of cells which, although sustaining a high proliferative and combined lympho-myeloid differentiation potential, have lost the ability to adopt erythroid and megakaryocyte lineage fates. Cells in the Lin-Sca-1+c-kit+ HSC compartment coexpressing high levels of the tyrosine kinase receptor Flt3 sustain granulocyte, monocyte, and B and T cell potentials but in contrast to Lin-Sca-1+c-kit+Flt3- HSCs fail to produce significant erythroid and megakaryocytic progeny. This distinct lineage restriction site is accompanied by downregulation of genes for regulators of erythroid and megakaryocyte development. In agreement with representing a lymphoid primed progenitor, Lin-Sca-1+c-kit+CD34+Flt3+ cells display upregulated IL-7 receptor gene expression. Based on these observations, we propose a revised road map for adult blood lineage development.
- Published
- 2005
- Full Text
- View/download PDF
33. IFN-γ Negatively Modulates Self-Renewal of Repopulating Human Hemopoietic Stem Cells
- Author
-
Sten Eirik W. Jacobsen, Liping Yang, Yutaka Sasaki, Ewa Sitnicka, David Bryder, Lennart Nilsson, and Ingunn Dybedal
- Subjects
Immunology ,Cell ,Dose-Response Relationship, Immunologic ,CD34 ,Antigens, CD34 ,Mice, SCID ,Biology ,Immunophenotyping ,Interferon-gamma ,Mice ,Antigens, CD ,Mice, Inbred NOD ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cell Lineage ,Progenitor cell ,ADP-ribosyl Cyclase ,Cells, Cultured ,Membrane Glycoproteins ,Hematopoietic Stem Cell Transplantation ,Cell Differentiation ,Fetal Blood ,Hematopoietic Stem Cells ,ADP-ribosyl Cyclase 1 ,Growth Inhibitors ,In vitro ,Clone Cells ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,Cord blood ,Stem cell - Abstract
Whereas multiple growth-promoting cytokines have been demonstrated to be involved in regulation of the hemopoietic stem cell (HSC) pool, the potential role of negative regulators is less clear. However, IFN-γ, if overexpressed, can mediate bone marrow suppression and has been directly implicated in a number of bone marrow failure syndromes, including graft-vs-host disease. Whether IFN-γ might directly affect the function of repopulating HSCs has, however, not been investigated. In the present study, we used in vitro conditions promoting self-renewing divisions of human HSCs to investigate the effect of IFN-γ on HSC maintenance and function. Although purified cord blood CD34+CD38− cells underwent cell divisions in the presence of IFN-γ, cycling HSCs exposed to IFN-γ in vitro were severely compromised in their ability to reconstitute long-term cultures in vitro and multilineage engraft NOD-SCID mice in vivo (>90% reduced activity in both HSC assays). In vitro studies suggested that IFN-γ accelerated differentiation of targeted human stem and progenitor cells. These results demonstrate that IFN-γ can negatively affect human HSC self-renewal.
- Published
- 2005
- Full Text
- View/download PDF
34. Human CD34+ hematopoietic stem cells capable of multilineage engrafting NOD/SCID mice express flt3: distinct flt3 and c-kit expression and response patterns on mouse and candidate human hematopoietic stem cells
- Author
-
Karina Liuba, Staffan Larsson, Ewa Sitnicka, Natalija Buza-Vidas, Jens M. Nygren, and Sten Erik W. Jacobsen
- Subjects
Cell Survival ,Genetic enhancement ,Transplantation, Heterologous ,Immunology ,CD34 ,Antigens, CD34 ,Mice, SCID ,Biology ,Biochemistry ,Mice ,Bone Marrow ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Progenitor cell ,Myelopoiesis ,Stem Cell Factor ,Severe combined immunodeficiency ,Lymphopoiesis ,Graft Survival ,Hematopoietic Stem Cell Transplantation ,Membrane Proteins ,Receptor Protein-Tyrosine Kinases ,hemic and immune systems ,Cell Biology ,Hematology ,Fetal Blood ,Hematopoietic Stem Cells ,medicine.disease ,Cell biology ,Proto-Oncogene Proteins c-kit ,Haematopoiesis ,medicine.anatomical_structure ,fms-Like Tyrosine Kinase 3 ,Cord blood ,embryonic structures ,Bone marrow ,Stem cell - Abstract
The cytokine tyrosine kinase receptors c-kit and flt3 are expressed and function in early mouse and human hematopoiesis. Through its ability to promote ex vivo expansion and oncoretroviral transduction of primitive human hematopoietic progenitors, the flt3 ligand (FL) has emerged as a key stimulator of candidate human hematopoietic stem cells (HSCs). However, recent studies in the mouse suggest that though it is present on short-term repopulating cells, flt3 is not expressed on bone marrow long-term reconstituting HSCs, the ultimate target for the development of cell replacement and gene therapy. Herein we demonstrate that though only a fraction of human adult bone marrow and cord blood CD34+long-term culture-initiating cells (LTC-ICs) express flt3, most cord blood lymphomyeloid HSCs capable of in vivo reconstituting nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice are flt3+. The striking difference in flt3 and c-kit expression on mouse and candidate human HSCs translated into a corresponding difference in flt3 and c-kit function because FL was more efficient than SCF at supporting the survival of candidate human HSCs. In contrast, SCF is far superior to FL as a viability factor for mouse HSCs. Thus, the present data provide compelling evidence for a contrasting expression and response pattern of flt3 and c-kit on mouse and human HSCs.
- Published
- 2003
- Full Text
- View/download PDF
35. A human ips model implicates embryonic B-myeloid fate restriction as a developmental susceptibility to ETV6-RUNX1
- Author
-
Stefan Karlsson, Emma Laycock, Javier Herrero, Ewa Sitnicka, John Brown, Jason Wray, Sten Erik W. Jacobsen, Charlotta Böiers, Tariq Enver, Virginia Turati, Dapeng Wang, Andrew D. M. Smith, Alya Zriwil, Chela James, and Simon Richardson
- Subjects
Genetics ,Cancer Research ,Myeloid ,medicine.anatomical_structure ,Etv6 runx1 ,medicine ,Cell Biology ,Hematology ,Biology ,Molecular Biology ,Embryonic stem cell - Published
- 2017
- Full Text
- View/download PDF
36. Resolving the fetal to adult switch in B lymphopoiesis at the single cell level
- Author
-
Stefan Lang, Trine A. Kristiansen, Shamit Soneji, Alya Zriwil, Stijn Vanhee, Ewa Sitnicka Quinn, David Bryder, Elin Jaensson-Gyllenbäck, Alexander Doyle, and Joan Yuan
- Subjects
Cancer Research ,Fetus ,B lymphopoiesis ,Genetics ,Cell Biology ,Hematology ,Biology ,Cellular level ,Molecular Biology ,Cell biology - Published
- 2017
- Full Text
- View/download PDF
37. Identification of a Human Natural Killer Cell Lineage-Restricted Progenitor in Fetal and Adult Tissues
- Author
-
Jakob Michaëlsson, Alya Zriwil, Claudia Peitzsch, Shamit Soneji, Virginie Renoux, Danielle Friberg, and Ewa Sitnicka
- Subjects
Adult ,Cytotoxicity, Immunologic ,Immunology ,Biology ,Natural killer cell ,Immunophenotyping ,Fetal Development ,Interleukin 21 ,Fetus ,Antigens, CD ,medicine ,Immunology and Allergy ,Humans ,Cell Lineage ,Lymphopoiesis ,Lymphokine-activated killer cell ,Clinical Laboratory Medicine ,Innate lymphoid cell ,Cell Differentiation ,Lymphoid Progenitor Cells ,Natural killer T cell ,Fetal Blood ,Immunity, Innate ,Cell biology ,Hematopoiesis ,Killer Cells, Natural ,Klinisk laboratoriemedicin ,Infectious Diseases ,medicine.anatomical_structure ,Interleukin 12 - Abstract
Natural killer (NK) cells are cytotoxic lymphocytes and play a vital role in controlling viral infections and cancer. In contrast to B and T lymphopoiesis where cellular and regulatory pathways have been extensively characterized, the cellular stages of early human NK cell commitment remain poorly understood. Here we demonstrate that a Lin(-)CD34(+)CD38(+)CD123(-)CD45RA(+)CD7(+)CD10(+)CD127(-) population represents a NK lineage-restricted progenitor (NKP) in fetal development, umbilical cord blood, and adult tissues. The newly identified NKP has robust NK cell potential both in vitro and in vivo, generates functionally cytotoxic NK cells, and lacks the ability to produce T cells, B cells, myeloid cells, and innate lymphoid-like cells (ILCs). Our findings identify an early step to human NK cell commitment and provide new insights into the human hematopoietic hierarchy.
- Published
- 2014
38. Junctional Adhesion Molecule 2 Represents a Novel Subset of Hematopoietic Stem Cells Poised for T Lymphopoiesis
- Author
-
Alya Zriwil, Mark van der Garde, Shin Kaneko, Valgardur Sigurdsson, Visnja Radulovic, Kenichi Miharada, Ewa Sitnicka Quinn, and Svetlana Soboleva
- Subjects
0301 basic medicine ,education.field_of_study ,T cell ,Immunology ,Population ,Cell Biology ,Hematology ,Cell cycle ,Biology ,Biochemistry ,Cell biology ,Transplantation ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,medicine ,Lymphopoiesis ,Stem cell ,education ,B cell - Abstract
Phenotypically well-characterized hematopoietic stem cells (HSCs) still represent a heterogeneous pool of primitive cells regarding to their functionality. In particular, different lineage potential of HSCs have been considered as one of key features of the HSC heterogeneity. The lineage output of HSCs is often coupled with cell cycle status or long-term reconstitution potential, however molecular mechanisms of the mutuality are unclear and other type of the regulation may exist. In addition, prospective isolation of such HSCs biased towards specific lineage(s) is still problematic, as many of categorizations highly rely on retrospective information, e.g. transplantation assay. Although several markers have been reported to be able to subdivide HSCs into subcategories, exploration of additional markers will allow us understanding further molecular mechanisms of HSC regulations including activation and lineage choice. Here, we show that cell surface expression of Junctional adhesion molecule 2 (Jam2) represents higher reconstitution capacity of HSCs and the T cell potential. Flow cytometry analyses revealed that a subset of CD150+CD48-KSL cells in mouse bone marrow (BM) were positive for Jam2 (Jam2+HSC, 36.6 ±13.0 %), while other Jam family member Jam1 (F11r) was expressed on all HSCs and Jam3 was not detected. To examine functional differences of Jam2+ and Jam2-HSCs, 30 cells were separately transplanted into lethally irradiated mice. Peripheral blood analyses revealed that Jam2+HSCs reconstituted more efficiently than Jam2-HSCs (77.5 ±15.9 and 51.7 ±29.3 %, respectively). In case of transplantation using 5 cells, the frequency of reconstituted mice was higher in Jam2+HSCs (7 in 11) compared to Jam2-HSCs (4 in 11), indicating that Jam2+ population is more enriched for functional HSCs. The expression of Jam2 on HSC is reversible, but not hierarchical, as both Jam2+ and Jam2-HSCs reconstituted opposite population in the BM.Lineage analyses revealed that Jam2+HSCs have a greater potential in lymphoid cell reconstitution, particularly T cells, whereas the chimerism in myeloid cells was not significantly different from Jam2-HSCs. This tendency of higher contribution to the T cell development was even more pronounced in the secondary transplantation experiments, where the contribution of Jam2+HSCs in T cells was close to 100 %. Of note, most of Jam2+HSCs were in a dormant state, suggesting that the T cell (or lymphoid) potential of Jam2+HSCs is independent of cell cycle progression. Jam2 has been reported to interact with Jam1, which mediates the Notch signaling (Kobayashi et al., Nature, 2014). Competitive co-culture of Jam2+ vs Jam2-HSCs on OP9-DL1 showed that Jam2+HSCs dominated the T cell production, whereas no difference was seen in B cell production upon OP9 co-culture. Since Jam2 positivity correlates to T cell potential, we asked if altered T lymphopoiesis environment affects the cell surface Jam2 expression. Comparison of C57BL/6, NOD, NOD-Scid and NOD-Scid Il2rγ KO (NSG) mice showed that HSCs of NSG mice have significantly higher frequency of Jam2+HSCs, suggesting that cell surface Jam2 expression might be regulated by specific cytokine(s) binding to IL2Rγ. Our findings suggest Jam2 is a new marker for a subset of HSCs that preferentially generate T cells. In addition, this work uncouples the lineage choice and cell cycle status, which proposes a novel model to the lineage-determining machineries. Since efficient and immediate generation of T cells in transplantation therapy is important to minimize infectious risks, understanding the molecular basis of the Jam-Notch cooperation would contribute to establish safer and more efficient treatment. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
- Full Text
- View/download PDF
39. Transforming growth factor beta 1 directly and reversibly inhibits the initial cell divisions of long-term repopulating hematopoietic stem cells
- Author
-
Gregory V. Priestley, Stephen H. Bartelmez, Ewa Sitnicka, Francis W. Ruscetti, and Norman S. Wolf
- Subjects
education.field_of_study ,Cell division ,Cell growth ,Cellular differentiation ,Immunology ,Population ,Stem cell factor ,Cell Biology ,Hematology ,Transforming growth factor beta ,Biology ,Biochemistry ,Cell biology ,Haematopoiesis ,biology.protein ,Stem cell ,education - Abstract
Hematopoiesis appears to be regulated, in part, by a balance between extracellular positive and negative growth signals. Transforming growth factor beta-1 (TGF-beta 1) has been shown to be a negative regulator of primitive hematopoietic cells. This study examined the direct effect of TGF-beta 1 on the proliferation and differentiation of long-term repopulating hematopoietic stem cells (LTR-HSC) in vitro. We previously reported a cell fractionation approach that includes the selection of low Hoescht 33342/low Rhodamine 123 (low Ho/Rh) cell fractions that are highly enriched for long-term repopulating cells (LTR-HSC) and also clone to a very high efficiency in the presence of stem cell factor (SCF) + interleukin-3 (IL-3) + IL-6: 90% to 100% of individually cultured low Ho/Rh cells formed high proliferative potential clones. This high cloning efficiency of an LTR-HSC enriched cell population enabled proliferation inhibition studies to be more easily interpreted. In this report, we show that the continuous presence of TGF-beta 1 directly inhibits the cell division of essentially all low Ho/Rh cells (in a dose-dependent manner) during their 0 to 5th cell division in vitro. Therefore, it follows that TGF-beta 1 must directly inhibit the proliferation of LTR-HSC contained within these low Ho/Rh cells. The time required for some low Ho/Rh cells to undergo their first cell division in vitro was also prolonged in the presence of TGF-beta 1. Furthermore, when low Ho/Rh cells were exposed to TFG-beta 1 for varying lengths of time before neutralization of the TGF-beta 1 by monoclonal antibody, the ability to form macroclones was markedly decreased after approximately 4 days of TGF-beta 1 exposure. In addition, 1 to 10 ng/mL of TGF-beta 1 resulted in a maintenance of high proliferative potential-colony-forming cell (HPP-CFC) during 8 days of culture compared with loss of HPP-CFC in cultures with no added TGF- beta 1. In conclusion, this study shows that TGF-beta 1 directly inhibits the initial stages of proliferation of LTR-HSC and appears to slow the differentiation of daughter cells of low Ho/Rh cells.
- Published
- 1996
- Full Text
- View/download PDF
40. The effect of thrombopoietin on the proliferation and differentiation of murine hematopoietic stem cells
- Author
-
Nan Lin, Gregory V. Priestley, Kenneth Kaushansky, Ewa Sitnicka, Virginia C. Broudy, Norman S. Wolf, and Norma E. Fox
- Subjects
Cellular differentiation ,Growth factor ,medicine.medical_treatment ,Immunology ,Stem cell factor ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Molecular biology ,Haematopoiesis ,medicine.anatomical_structure ,Megakaryocyte ,medicine ,Stem cell ,Thrombopoietin ,Interleukin 3 - Abstract
In this study, we explored whether thrombopoietin (Tpo) has a direct in vitro effect on the proliferation and differentiation of long-term repopulating hematopoietic stem cells (LTR-HSC). We previously reported a cell separation method that uses the fluorescence-activated cell sorter selection of low Hoescht 33342/low Rhodamine 123 (low Ho/low Rh) fluorescence cell fractions that are highly enriched for LTR-HSC and can reconstitute lethally irradiated recipients with fewer than 20 cells. Low Ho/low Rh cells clone with high proliferative potential in vitro in the presence of stem cell factor (SCF) + interleukin-3 (IL-3) + IL-6 (90% to 100% HPP-CFC). Tpo alone did not induce proliferation of these low Ho/low Rh cells. However, in combination with SCF or IL-3, Tpo had several synergistic effects on cell proliferation. When Tpo was added to single growth factors (either SCF or IL-3 or the combination of both), the time required for the first cell division of low Ho/low Rh cells was significantly shortened and their cloning efficiency increased substantially. Moreover, the subsequent clonal expansion at the early time points of culture was significantly augmented by Tpo. Low Ho/low Rh cells, when assayed in agar directly after sorting, did not form megakaryocyte colonies in any growth condition tested. Several days of culture in the presence of multiple cytokines were required to obtain colony-forming units-megakaryocyte (CFU-Mk). In contrast, more differentiated, low Ho/high Rh cells, previously shown to contain short- term repopulating hematopoietic stem cells (STR-HSC), were able to form megakaryocyte colonies in agar when cultured in Tpo alone directly after sorting. These data establish that Tpo acts directly on primitive hematopoietic stem cells selected using the Ho/Rh method, but this effect is dependent on the presence of pluripotent cytokines. These cells subsequently differentiate into CFU-Mk, which are capable of responding to Tpo alone. Together with the results of previous reports of its effects on erythroid progenitors, these results suggest that the effects of Tpo on hematopoiesis are greater than initially anticipated.
- Published
- 1996
- Full Text
- View/download PDF
41. Do the Preclinical Effects of Thrombopoietin Correlate with Its In Vitro Properties?
- Author
-
K. Sprugel, Catherine Lofton-Day, Kenneth Kaushansky, A. Grossman, Virginia C. Broudy, and Ewa Sitnicka
- Subjects
Erythrocytes ,Bone Marrow Cells ,Biology ,Blood cell ,Mice ,Megakaryocyte ,Granulocyte Colony-Stimulating Factor ,medicine ,Animals ,Humans ,Progenitor cell ,Erythropoietin ,Thrombopoietin ,Interleukin 3 ,Stem Cells ,Cell Biology ,Haematopoiesis ,medicine.anatomical_structure ,Immunology ,Molecular Medicine ,Erythropoiesis ,Stem cell ,Megakaryocytes ,Cell Division ,Spleen ,Granulocytes ,Developmental Biology - Abstract
In the short time since its cloning, much has been learned of the in vitro properties of thrombopoietin (TPO). In addition to effects on the differentiation of megakaryocytes, TPO has also been shown to stimulate the proliferation of megakaryocytic progenitor cells, colony-forming units-megakaryocytes (CFU-MK), to act in synergy with interleukin 3 or c-kit ligand and erythropoietin (Epo) to stimulate the development of early and the generation of late erythroid progenitor cells, and to affect the rate of entry into the cell cycle and proliferative capacity of hematopoietic stem cells. An important question posed by these observations, for both TPO and for hematopoietic research in general, is whether the in vitro effects of a cytokine are mirrored by its preclinical and clinical biology. The results of recent studies in mice and nonhuman primates will be presented which have attempted to address this issue. In normal animals, TPO increases the numbers of marrow and spleen CFU-granulocyte/erythroid/macrophage/megakaryocyte, CFU-MK, CFU-GM and BFU-E, but its effects in the peripheral blood are limited to marked increases in the platelet count. The reason for widespread progenitor cell effects, yet stable leukocyte and erythrocyte blood counts, is likely the predominant regulatory effects of G-CSF and Epo; in the absence of elevated levels of the lineage-dominant regulator of each of these cell types, expanded progenitor cell numbers are not translated into increased peripheral blood counts. However, in states of increased blood cell demand such as follows myelosuppressive therapy, elevated levels of Epo and G-CSF allow the effects of TPO on erythropoiesis and myelopoiesis to become manifest. The administration of TPO to myelosuppressed animals is associated with not only greatly expanded hematopoietic progenitor cell recovery, but also improvement in platelet, red cell and leukocyte nadir levels and greatly accelerated recovery of all three cell lineages. These results indicate that the panhematopoietic properties of TPO identified by in vitro culture techniques correlate well with its effects in animals. The results of ongoing clinical trials should soon establish whether these conclusions can be extended to patient care.
- Published
- 1996
- Full Text
- View/download PDF
42. Support versus inhibition of hematopoiesis by two characterized stromal cell types
- Author
-
Qi Ru Wang, Schickwann Tsai, Ewa Sitnicka, and Norman S. Wolf
- Subjects
Male ,Stromal cell ,Stem cell factor ,Cell Communication ,Biology ,Mice ,Lymph node stromal cell ,medicine ,Animals ,Cell Lineage ,Cell Differentiation ,Cell Biology ,Fibroblasts ,Alkaline Phosphatase ,Hematopoiesis ,Cell biology ,Mice, Inbred C57BL ,Endothelial stem cell ,Haematopoiesis ,medicine.anatomical_structure ,Mice, Inbred DBA ,Immunology ,Molecular Medicine ,Female ,Endothelium, Vascular ,Bone marrow ,Stromal Cells ,Stem cell ,Cell Division ,Developmental Biology ,Adult stem cell - Abstract
Stromal cells are believed to regulate hematopoiesis through direct cell-cell contact interactions and the release of growth factors. Many questions remain, however, about their lineage derivation and functional heterogeneity. We have previously shown that the adherent nontransformed, nonimmortalized murine bone marrow stromal cell population consists of three cell types which could be grown separately in vitro. Based on the phenotype characterization and expression of surface antigens, we proposed a classification listing for murine bone marrow stromal cells as macrophages, endothelial-like cells and myofibroblasts that display smooth muscle-like characteristics in culture. The present study describes the ability of each of these freshly isolated separated murine stromal cell populations to support the growth of primitive hematopoietic stem cells previously characterized as highly enriched in long-term repopulating cells (LTRC). Of the three stromal cell types tested only the myofibroblasts were capable of support for multilineage hematopoiesis derived in vitro from LTRC in a cloning ring culture system. Endothelial-like cells had an inhibitory effect on the proliferation of LTRC and their descendant cells that was induced by exogenous growth factors. This inhibitory activity was present in a low molecular weight filtrate of endothelial-like cells culture medium. This suggests an essential role for marrow stroma myofibroblasts in the support of proliferation of hematopoietic cells at the stage of early divisions of primitive hematopoietic stem cells and endothelial-like cells as negative regulators of this proliferation.
- Published
- 1995
- Full Text
- View/download PDF
43. Distinct and overlapping direct effects of macrophage inflammatory protein-1 alpha and transforming growth factor beta on hematopoietic progenitor/stem cell growth
- Author
-
Stephen H. Bartelmez, Francis W. Ruscetti, Jonathan R. Keller, Sten Eirik W. Jacobsen, John Gooya, Mariaestella Ortiz, and Ewa Sitnicka
- Subjects
TGF alpha ,Immunology ,Cell Biology ,Hematology ,Transforming growth factor beta ,Endoglin ,Biology ,Biochemistry ,Molecular biology ,Haematopoiesis ,Transforming growth factor, beta 3 ,TGF beta signaling pathway ,biology.protein ,Stem cell ,Progenitor cell - Abstract
Both transforming growth factor beta (TGF beta) and macrophage inflammatory protein 1 alpha (MIP-1 alpha) have been shown to be multifunctional regulators of hematopoiesis that can either inhibit or enhance the growth of hematopoietic progenitor cells (HPC). We report here the spectrum of activities of these two cytokines on different hematopoietic progenitor and stem cell populations, and whether these effects are direct or indirect. MIP-1 alpha enhances interleukin-3 (IL- 3)/and granulocyte-macrophage colony-stimulating factor (GM- CSF)/induced colony formation of normal bone marrow progenitor cells (BMC) and lineage-negative (Lin-) progenitors, but has no effect on G- CSF or CSF-1/induced colony formation. Similarly, TGF beta enhances GM- CSF/induced colony formation of normal BMC and Lin- progenitors. In contrast, TGF beta inhibits IL-3/ and CSF-1/induced colony formation of Lin- progenitors. The effects of MIP-1 alpha and TGF beta on the growth of Lin- progenitors were direct and correlate with colony formation in soft agar. Separation of the Lin- cells into Thy-1 and Thy-1lo subsets showed that the growth of Thy-1lo Lin- cells is directly inhibited by MIP-1 alpha and TGF beta regardless of the cytokine used to stimulate growth (IL-3), GM-CSF, or CSF-1). In contrast, two other stem cell populations (0% to 15% Hoechst 33342/Rhodamine 123 [Ho/Rh123] and Lin- Sca-1+ cells) were markedly inhibited by TGF beta and unaffected by MIP- 1 alpha. Furthermore, MIP-1 alpha has no effect on high proliferative potential colony-forming cells 1 or 2 (HPP-CFC/1 or /2) colony formation in vitro, whereas TGF beta inhibits both HPP-CFC/1 and HPP- CFC/2. Thus, MIP-1 alpha and TGF beta are direct bidirectional regulators of HPC growth, whose effects are dependent on other growth factors present as well as the maturational state of the HPC assayed. The spectrum of their inhibitory and enhancing activities shows overlapping yet distinct effects.
- Published
- 1994
- Full Text
- View/download PDF
44. Emergence of NK-cell progenitors and functionally competent NK-cell lineage subsets in the early mouse embryo
- Author
-
Min Cheng, Hojjatollah Nozad Charoudeh, Sten Eirik W. Jacobsen, Ewa Sitnicka, P. Chaves, Yanjuan Tang, and Claudia Peitzsch
- Subjects
T cell ,T-Lymphocytes ,Immunology ,Thymus Gland ,Biology ,Biochemistry ,Interleukin 21 ,Mice ,Pregnancy ,medicine ,Cytotoxic T cell ,Animals ,Antigens, Ly ,Cell Lineage ,Myeloid Cells ,Lymphopoiesis ,Progenitor cell ,Cells, Cultured ,B-Lymphocytes ,Lymphokine-activated killer cell ,Cell growth ,Stem Cells ,Cell Differentiation ,Cell Biology ,Hematology ,Embryonic stem cell ,Interleukin-2 Receptor beta Subunit ,Killer Cells, Natural ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Liver ,Immune System ,embryonic structures ,Female ,Stromal Cells ,Spleen ,NK Cell Lectin-Like Receptor Subfamily B - Abstract
The earliest stages of natural killer (NK)–cell development are not well characterized. In this study, we investigated in different fetal hematopoietic tissues how NK-cell progenitors and their mature NK-cell progeny emerge and expand during fetal development. Here we demonstrate, for the first time, that the counterpart of adult BM Lin−CD122+NK1.1−DX5− NK-cell progenitor (NKP) emerges in the fetal liver at E13.5. After NKP expansion, immature NK cells emerge at E14.5 in the liver and E15.5 in the spleen. Thymic NK cells arise at E15.5, whereas functionally competent cytotoxic NK cells were present in the liver and spleen at E16.5 and E17.5, respectively. Fetal NKPs failed to produce B and myeloid cells but sustained combined NK- and T-lineage potential at the single-cell level. NKPs were also found in the fetal blood, spleen, and thymus. These findings show the emergence and expansion of bipotent NK/T-cell progenitor during fetal and adult lymphopoiesis, further supporting that NK/T-lineage restriction is taking place prethymically. Uncovering the earliest NK-cell developmental stages will provide important clues, helping to understand the origin of diverse NK-cell subsets, their progenitors, and key regulators.
- Published
- 2011
45. Impact of gene dosage, loss of wild-type allele, and FLT3 ligand on Flt3-ITD-induced myeloproliferation
- Author
-
Claus Nerlov, Anne Hultquist, Shabnam Kharazi, D Atkinson, Adam J. Mead, Kristian Reckzeh, Ewa Sitnicka, Toshio Suda, Fumio Arai, Lars Rönnstrand, Natalija Buza-Vidas, Sidinh Luc, Charlotta Böiers, Zhi Ma, Kristina Masson, Anna Mansour, Jörg Cammenga, Helen Ferry, and Sten Eirik W. Jacobsen
- Subjects
Male ,Myeloid ,Immunology ,Gene Dosage ,Loss of Heterozygosity ,Bone Marrow Cells ,Mice, Transgenic ,Biology ,Biochemistry ,Gene dosage ,Mice ,fluids and secretions ,Growth factor receptor ,Gene Duplication ,hemic and lymphatic diseases ,Myeloproliferation ,medicine ,Animals ,Gene Knock-In Techniques ,Allele ,Autocrine signalling ,Alleles ,Cells, Cultured ,Cell Proliferation ,Myeloproliferative Disorders ,Wild type ,Membrane Proteins ,Myeloid leukemia ,hemic and immune systems ,Cell Biology ,Hematology ,Molecular biology ,Mice, Inbred C57BL ,Phenotype ,medicine.anatomical_structure ,fms-Like Tyrosine Kinase 3 ,Tandem Repeat Sequences ,embryonic structures ,psychological phenomena and processes - Abstract
Acquisition of homozygous activating growth factor receptor mutations might accelerate cancer progression through a simple gene-dosage effect. Internal tandem duplications (ITDs) of FLT3 occur in approximately 25% cases of acute myeloid leukemia and induce ligand-independent constitutive signaling. Homozygous FLT3-ITDs confer an adverse prognosis and are frequently detected at relapse. Using a mouse knockin model of Flt3–internal tandem duplication (Flt3-ITD)–induced myeloproliferation, we herein demonstrate that the enhanced myeloid phenotype and expansion of granulocyte-monocyte and primitive Lin−Sca1+c-Kit+ progenitors in Flt3-ITD homozygous mice can in part be mediated through the loss of the second wild-type allele. Further, whereas autocrine FLT3 ligand production has been implicated in FLT3-ITD myeloid malignancies and resistance to FLT3 inhibitors, we demonstrate here that the mouse Flt3ITD/ITD myeloid phenotype is FLT3 ligand-independent.
- Published
- 2011
46. Dissecting the hematopoietic microenvironment. IX. Further characterization of murine bone marrow stromal cells
- Author
-
Rui-Gao Fei, Norman S. Wolf, Ewa Sitnicka, Philip E. Penn, and DeZhao Jiang
- Subjects
Pathology ,medicine.medical_specialty ,Cell type ,Stromal cell ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Molecular biology ,medicine.anatomical_structure ,Cell culture ,Myosin ,medicine ,Macrophage ,Bone marrow ,Fibroblast ,Fetal bovine serum - Abstract
We have previously shown the adherent nontransformed, nonimmortalized murine bone marrow stromal cell (BMSC) population to consist of phagocytic macrophage and endothelial-like cells and nonphagocytic fibroblasts. Both colonial and near confluent growth of each cell type was obtained following magnetic bead separation, subsequent passaging, and sustained culture with fetal bovine serum and cytokines. Monoclonal antibody staining of antigenic determinants was used to characterize the phenotype of the stromal cell population in primary platings of murine colony-forming unit fibroblast and long-term bone marrow cultures. The antibodies MECA-99, MECA-32, and MJ7–18, raised against murine vascular endothelial antigenic determinants, and von Willebrand's factor all stained selectively for the rounded endothelial- like cells. Endothelial-like cells as well as macrophages expressed the myeloid surface antigens F4/80, 7/4, and Mac-1 under our culture conditions. The cytoskeleton of the stromal fibroblasts in culture was shown to express smooth muscle-specific actin isoforms, as evidenced by positive staining of stress fibers for alpha smooth muscle-1, CGA-7 (alpha/gamma isoforms), and HHF-35 (recognizes all muscle-specific actins). Under culture conditions, stromal fibroblasts were also found to be positive for a polyclonal smooth muscle myosin. It was found that these fibroblasts stained for collagens type I, III, and IV in our cultures. Although collagen type IV is considered a by-product of endothelial cells, endothelial-like cells in our cultures did not stain for any of the collagen types. We propose a classification listing for murine BMSCs as macrophages, endotheliallike cells, and fibroblasts that display smooth muscle-like characteristics in culture.
- Published
- 1993
- Full Text
- View/download PDF
47. Early cellular pathways of mouse natural killer cell development
- Author
-
Ewa Sitnicka
- Subjects
Innate immune system ,Lymphokine-activated killer cell ,Cellular differentiation ,Hematopoietic stem cell ,Immunology in the medical area ,Cell Differentiation ,Biology ,Natural killer T cell ,Hematopoietic Stem Cells ,Cell biology ,Transplantation ,Killer Cells, Natural ,Mice ,medicine.anatomical_structure ,Immunology ,medicine ,Immunology and Allergy ,Animals ,Cell Lineage ,Bone marrow ,Progenitor cell - Abstract
Natural killer (NK) cells are large granular lymphocytes that are components of the innate immune system. These cells are key players in the defense against viral and other microbial infections and cancer and have an important function during pregnancy, autoimmunity and allergy. Furthermore, NK cells play important roles in hematopoietic stem cell (HSC) transplantation by providing the graft versus leukemia effect and preventing the development of graft versus host disease. Thus, understanding the developmental pathway(s) from multipotent HSCs to the NK cell lineage-restricted progenitors is of significant clinical value. However, despite extensive progress in the delineation of mature blood cell development, including the B- and T-cell lineages, the early stages of NK cell lineage commitment and development have been less well established and characterized. Here, I review the progress made thus far in dissecting the developmental stages, from HSCs in the bone marrow to the lineage-committed NK cells in mouse.
- Published
- 2010
48. Expression and role of FLT3 in regulation of the earliest stage of normal granulocyte-monocyte progenitor development
- Author
-
Christina Jensen, Anne Hultquist, Lilian Wittmann, Shabnam Kharazi, Sten Eirik W. Jacobsen, Cornelis J.H. Pronk, Ewa Sitnicka, Natalija Buza-Vidas, and Charlotta Böiers
- Subjects
Myeloid ,Immunology ,Gene Expression ,Biology ,Biochemistry ,Monocytes ,Mice ,fluids and secretions ,hemic and lymphatic diseases ,medicine ,Animals ,Cell Lineage ,Myeloid Cells ,Progenitor cell ,Thrombopoietin ,Progenitor ,Mice, Knockout ,Myeloid leukemia ,Membrane Proteins ,hemic and immune systems ,Cell Biology ,Hematology ,medicine.disease ,Hematopoietic Stem Cells ,Phenotype ,Mice, Inbred C57BL ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,fms-Like Tyrosine Kinase 3 ,Fms-Like Tyrosine Kinase 3 ,embryonic structures ,Cancer research ,Granulocytes ,Signal Transduction - Abstract
Mice deficient in c-fms–like tyrosine kinase 3 (FLT3) signaling have reductions in early multipotent and lymphoid progenitors, whereas no evident myeloid phenotype has been reported. However, activating mutations of Flt3 are among the most common genetic events in acute myeloid leukemia and mice harboring internal tandem duplications within Flt3 (Flt3-ITD) develop myeloproliferative disease, with characteristic expansion of granulocyte-monocyte (GM) progenitors (GMP), possibly compatible with FLT3-ITD promoting a myeloid fate of multipotent progenitors. Alternatively, FLT3 might be expressed at the earliest stages of GM development. Herein, we investigated the expression, function, and role of FLT3 in recently identified early GMPs. Flt3-cre fate-mapping established that most progenitors and mature progeny of the GM lineage are derived from Flt3-expressing progenitors. A higher expression of FLT3 was found in preGMP compared with GMP, and preGMPs were more responsive to stimulation with FLT3 ligand (FL). Whereas preGMPs and GMPs were reduced in Fl−/− mice, megakaryocyte-erythroid progenitors were unaffected and lacked FLT3 expression. Notably, mice deficient in both thrombopoietin (THPO) and FL had a more pronounced GMP phenotype than Thpo−/− mice, establishing a role of FL in THPO-dependent and -independent regulation of GMPs, of likely significance for myeloid malignancies with Flt3-ITD mutations.
- Published
- 2010
49. FLT3 receptor and ligand are dispensable for maintenance and posttransplantation expansion of mouse hematopoietic stem cells
- Author
-
Min Cheng, Ewa Sitnicka, Natalija Buza-Vidas, Sara Duarte, Hojjatollah Nozad Charoudeh, and Sten Eirik W. Jacobsen
- Subjects
Immunology ,Regulator ,Biology ,Biochemistry ,Mice ,hemic and lymphatic diseases ,medicine ,Animals ,Receptor ,Graft Survival ,Hematopoietic Stem Cell Transplantation ,Hematopoietic stem cell ,Membrane Proteins ,hemic and immune systems ,Cell Biology ,Hematology ,Flow Cytometry ,Hematopoietic Stem Cells ,Mice, Mutant Strains ,Cell biology ,Hematopoiesis ,Transplantation ,Mice, Inbred C57BL ,Haematopoiesis ,medicine.anatomical_structure ,Phenotype ,fms-Like Tyrosine Kinase 3 ,Fms-Like Tyrosine Kinase 3 ,embryonic structures ,Stem cell ,Ex vivo ,Cell Division - Abstract
Originally cloned from hematopoietic stem cell (HSC) populations and its ligand being extensively used to promote ex vivo HSC expansion, the FMS-like tyrosine kinase 3 (FLT3; also called FLK2) receptor and its ligand (FL) were expected to emerge as an important physiologic regulator of HSC maintenance and expansion. However, the role of FLT3 receptor and ligand in HSC regulation remains unclear and disputed. Herein, using Fl-deficient mice, we establish for the first time that HSC expansion in fetal liver and after transplantation is FL independent. Because previous findings in Flk2−/− mice were compatible with an important role of FLT3 receptor in HSC regulation and because alternative ligands might potentially interact directly or indirectly with FLT3 receptor, we here also characterized HSCs in Flk2−/− mice. Advanced phenotypic as well as functional evaluation of Flk2−/− HSCs showed that the FLT3 receptor is dispensable for HSC steady-state maintenance and expansion after transplantation. Taken together, these studies show that the FLT3 receptor and ligand are not critical regulators of mouse HSCs, neither in steady state nor during fetal or posttransplantation expansion.
- Published
- 2009
50. FLT3 ligand and not TSLP is the key regulator of IL-7-independent B-1 and B-2 B lymphopoiesis
- Author
-
Min Cheng, Shabnam Kharazi, Sten Eirik W. Jacobsen, Ewa Sitnicka, Anna Lübking, Christina Jensen, and Charlotta Böiers
- Subjects
Thymic stromal lymphopoietin ,medicine.medical_treatment ,Immunology ,Cell ,Regulator ,Biology ,Biochemistry ,Mice ,Fetus ,Thymic Stromal Lymphopoietin ,medicine ,Animals ,Cell Lineage ,Lymphopoiesis ,Progenitor cell ,Progenitor ,B-Lymphocytes ,Interleukin-7 ,Interleukin ,Membrane Proteins ,Cell Biology ,Hematology ,Cell biology ,Cytokine ,medicine.anatomical_structure ,Cytokines - Abstract
Phenotypically and functionally distinct progenitors and developmental pathways have been proposed to exist for fetally derived B-1 and conventional B-2 cells. Although IL-7 appears to be the primary cytokine regulator of fetal and adult B lymphopoiesis in mice, considerable fetal B lymphopoiesis and postnatal B cells are sustained in the absence of IL-7; in humans, B-cell generation is suggested to be largely IL-7–independent, as severe combined immune-deficient patients with IL-7 deficiency appear to have normal B-cell numbers. However, the role of other cytokines in IL-7–independent B lymphopoiesis remains to be established. Although thymic stromal lymphopoietin (TSLP) has been proposed to be the main factor driving IL-7–independent B lymphopoiesis and to distinguish fetal from adult B-cell progenitor development in mice, recent studies failed to support a primary role of TSLP in IL-7–independent fetal B-cell development. However, the role of TSLP in IL-7–independent adult B lymphopoiesis and in particular in regulation of B-1 cells remains to be established. Here we demonstrate that, rather than TSLP, IL-7 and FLT3 ligand are combined responsible for all B-cell generation in mice, including recently identified B-1–specified cell progenitors. Thus, the same IL-7– and FLT3 ligand–mediated signal-ing regulates alternative pathways of fetal and adult B-1 and B-2 lymphopoiesis.
- Published
- 2008
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.