Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Physiol Cell Physiol ; 320(4): C509-C519, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33406026

ABSTRACT

Lysophosphatidic acid (LPA) is one of the lipids identified to be involved in stem cell differentiation. It exerts various functions through activation of G protein-coupled lysophosphatidic acid receptors (LPARs). In previous studies, we have demonstrated that activation of LPA receptor 3 (LPA3) promotes erythropoiesis of human hematopoietic stem cells (HSCs) and zebrafish using molecular and pharmacological approaches. Our results show that treatment with lysophosphatidic acid receptor 2 (LPA2) agonist suppressed erythropoiesis, whereas activation of LPA3 by 1-oleoyl-2-methyl-sn-glycero-3-phosphothionate (2S-OMPT) promoted it, both in vitro and in vivo. Furthermore, we have demonstrated the inhibitory role of LPA3 during megakaryopoiesis. However, the mechanism underlying these observations remains elusive. In the present study, we suggest that the expression pattern of LPARs may be correlated with the transcriptional factors GATA-1 and GATA-2 at different stages of myeloid progenitors. We determined that manipulation of GATA factors affected the expression levels of LPA2 and LPA3 in K562 leukemia cells. Using luciferase assays, we demonstrate that the promoter regions of LPAR2 and LPAR3 genes were regulated by these GATA factors in HEK293T cells. Mutation of GATA-binding sites in these regions abrogated luciferase activity, suggesting that LPA2 and LPA3 are regulated by GATA factors. Moreover, physical interaction between GATA factors and the promoter region of LPAR genes was verified in K562 cells using chromatin immunoprecipitation (ChIP) studies. Taken together, our results suggest that balance between LPA2 and LPA3 expression, which may be determined by GATA factors, is a regulatory switch for lineage commitment in myeloid progenitors. The expression-level balance of LPA receptor subtypes represents a novel mechanism regulating erythropoiesis and megakaryopoiesis.


Subject(s)
Cell Lineage , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Transcription, Genetic , Binding Sites , Erythropoiesis , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , HEK293 Cells , Humans , K562 Cells , Promoter Regions, Genetic , Receptors, Lysophosphatidic Acid/genetics , Signal Transduction , Thrombopoiesis
2.
Article in English | MEDLINE | ID: mdl-33035680

ABSTRACT

Hematopoiesis, the complex developmental process that forms blood components and replenishes the blood system, involves multiple intracellular and extracellular mechanisms. We previously demonstrated that lysophosphatidic acid (LPA), a lipid growth factor, has opposing regulatory effects on erythrocyte differentiation through activation of LPA receptors 2 and 3; yet the mechanisms underlying this process remain unclear. In this study, LPA2 is observed that highly expressed in common myeloid progenitors (CMP) in murine myeloid cells, whereas the expression of LPA3 displaces in megakaryocyte-erythroid progenitors (MEP) of later stage of myeloid differentiation. Therefore, we hypothesized that the switching expression of LPA2 and LPA3 determine the hematic homeostasis of mammalian megakaryocytic-erythroid lineage. In vitro colony-forming unit assays of murine progenitors reveal that LPA2 agonist GRI reduces the erythroblast differentiation potential of CMP. In contrast, LPA3 agonist OMPT increases the production of erythrocytes from megakaryocyte-erythrocyte progenitor cells (MEP). In addition, treatment with GRI reduces the erythroid, CMP, and MEP populations in mice, indicating that LPA2 predominantly inhibits myeloid differentiation at an early stage. In contrast, activation of LPA3 increases the production of terminally differentiated erythroid cells through activation of erythropoietic transcriptional factor. We also demonstrate that the LPA3 signaling is essential for restoration of phenylhydrazine (PHZ)-induced acute hemolytic anemia in mice and correlates to erythropoiesis impairment of Hutchinson-Gilford progeria Symptom (HGPS) premature aging expressed K562 model. Our results reveal the distinct roles of LPA2 and LPA3 at different stages of hematopoiesis in vivo, providing potentiated therapeutic strategies of anemia treatment.


Subject(s)
Anemia, Hemolytic/genetics , Erythroid Cells/metabolism , Erythropoiesis/genetics , Myeloid Cells/metabolism , Receptors, Lysophosphatidic Acid/genetics , Stem Cells/metabolism , Anemia, Hemolytic/chemically induced , Anemia, Hemolytic/drug therapy , Anemia, Hemolytic/metabolism , Animals , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Lineage/genetics , Disease Models, Animal , Erythroid Cells/cytology , Erythroid Cells/drug effects , Erythropoiesis/drug effects , Gene Expression Regulation , Humans , Isoquinolines/pharmacology , K562 Cells , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred BALB C , Myeloid Cells/cytology , Myeloid Cells/drug effects , Organothiophosphates/pharmacology , Phenylhydrazines/administration & dosage , Phosphatidic Acids/pharmacology , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/metabolism , Stem Cells/cytology , Stem Cells/drug effects
3.
Int J Mol Sci ; 21(6)2020 Mar 16.
Article in English | MEDLINE | ID: mdl-32188052

ABSTRACT

Vertebrate hematopoiesis is a complex physiological process that is tightly regulated by intracellular signaling and extracellular microenvironment. In recent decades, breakthroughs in lineage-tracing technologies and lipidomics have revealed the existence of numerous lipid molecules in hematopoietic microenvironment. Lysophosphatidic acid (LPA), a bioactive phospholipid molecule, is one of the identified lipids that participates in hematopoiesis. LPA exhibits various physiological functions through activation of G-protein-coupled receptors. The functions of these LPARs have been widely studied in stem cells, while the roles of LPARs in hematopoietic stem cells have rarely been examined. Nonetheless, mounting evidence supports the importance of the LPA-LPAR axis in hematopoiesis. In this article, we have reviewed regulation of hematopoiesis in general and focused on the microenvironmental and intracellular effects of the LPA in hematopoiesis. Discoveries in these areas may be beneficial to our understanding of blood-related disorders, especially in the context of prevention and therapy for anemia.


Subject(s)
Cellular Microenvironment/physiology , Hematopoiesis/physiology , Lysophospholipids/metabolism , Signal Transduction/physiology , Anemia/metabolism , Animals , Hematopoietic Stem Cells/physiology , Humans , Lipidomics , Receptors, Lysophosphatidic Acid , Transcription Factors
4.
Haematologica ; 105(1): 38-46, 2020 01.
Article in English | MEDLINE | ID: mdl-31806690

ABSTRACT

Hematopoietic stem cell (HSC) aging was originally thought to be essentially an HSC-autonomous process, which is the focus of another review in the same issue of Haematologica However, studies on the microenvironment that maintains and regulates HSC (HSC niche) over the past 20 years have suggested that microenvironmental aging contributes to declined HSC function over time. The HSC niches comprise a complex and dynamic molecular network of interactions across multiple cell types, including endothelial cells, mesenchymal stromal cells, osteoblasts, adipocytes, neuroglial cells and mature hematopoietic cells. Upon aging, functional changes in the HSC niches, such as microenvironmental senescence, imbalanced bone marrow mesenchymal stromal cell differentiation, vascular remodeling, changes in adrenergic signaling and inflammation, coordinately and dynamically influence the fate of HSC and their downstream progeny. The end result is lymphoid deficiency and myeloid skewing. During this process, aged HSC and their derivatives remodel the niche to favor myeloid expansion. Therefore, the crosstalk between HSC and the microenvironment is indispensable for the aging of the hematopoietic system and might represent a therapeutic target in age-related pathological disorders.


Subject(s)
Endothelial Cells , Hematopoietic Stem Cells , Cell Differentiation , Stem Cell Niche
5.
Cell Stem Cell ; 25(3): 407-418.e6, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31303548

ABSTRACT

Hematopoietic stem cells (HSCs) residing in the bone marrow (BM) accumulate during aging but are functionally impaired. However, the role of HSC-intrinsic and -extrinsic aging mechanisms remains debated. Megakaryocytes promote quiescence of neighboring HSCs. Nonetheless, whether megakaryocyte-HSC interactions change during pathological/natural aging is unclear. Premature aging in Hutchinson-Gilford progeria syndrome recapitulates physiological aging features, but whether these arise from altered stem or niche cells is unknown. Here, we show that the BM microenvironment promotes myelopoiesis in premature/physiological aging. During physiological aging, HSC-supporting niches decrease near bone but expand further from bone. Increased BM noradrenergic innervation promotes ß2-adrenergic-receptor(AR)-interleukin-6-dependent megakaryopoiesis. Reduced ß3-AR-Nos1 activity correlates with decreased endosteal niches and megakaryocyte apposition to sinusoids. However, chronic treatment of progeroid mice with ß3-AR agonist decreases premature myeloid and HSC expansion and restores the proximal association of HSCs to megakaryocytes. Therefore, normal/premature aging of BM niches promotes myeloid expansion and can be improved by targeting the microenvironment.


Subject(s)
Aging, Premature/pathology , Aging/physiology , Bone Marrow/physiology , Hematopoietic Stem Cells/physiology , Megakaryocytes/physiology , Myeloid Cells/physiology , Progeria/pathology , Adrenergic Agonists/administration & dosage , Aging/metabolism , Aging, Premature/metabolism , Animals , Cell Differentiation , Cell Encapsulation , Cell Proliferation , Disease Models, Animal , Humans , Interleukin-6/metabolism , Mice , Nitric Oxide Synthase Type I/metabolism , Progeria/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Stem Cell Niche
6.
Stem Cells Dev ; 27(3): 216-224, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29239275

ABSTRACT

Lysophosphatidic acid (LPA) is a membrane-derived lysophospholipid that exists in the plasma and platelets. It exerts its functions through activation of various LPA receptors (LPARs), which belong to the family of G protein-coupled receptors. Activation of LPARs has important roles in stem cell differentiation. However, how LPA affects human hematopoietic stem cell (HSC) differentiation remains elusive. In our previous studies, we have suggested that LPA receptor 2 (LPA2) and LPA receptor 3 (LPA3) play opposing roles and may act as a molecular switch during megakaryocytic differentiation in K562 cells. In this study, human CD34+ HSCs and zebrafish are adopted to investigate the roles of LPA3 during megakaryopoiesis/thrombopoiesis in vitro and in vivo. Our results show that LPAR3 mRNA expression level is decreased upon induction by thrombopoietin and stem cell factor in human HSCs. Using pharmacological activators and shRNA knockdown experiments, we demonstrate that activation of LPA3 inhibits megakaryopoiesis in human HSCs. In addition, pharmacological activation of LPA3 suppressed thrombopoiesis in zebrafish. Furthermore, blockage of LPA3 translation by morpholino increased the number of CD41-GFP+ cells in Tg(CD41:eGFP) zebrafish. Moreover, the mRNA expression level of zCD41 increased significantly in LPA3-knockout zebrafish. These results clarify the negative role of LPA3 during megakaryopoiesis and provide important information for potential treatments of related diseases, such as megakaryopenia.


Subject(s)
Hematopoietic Stem Cells/metabolism , Megakaryocytes/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Thrombopoiesis , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Humans , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
7.
Sci Rep ; 7(1): 11212, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28894229

ABSTRACT

Neuroblastoma (NB) is a childhood cancer with a low survival rate and great metastatic potential. Vascular endothelial growth factor (VEGF), an angiogenesis factor, has been found to be involved in CRT-related neuronal differentiation of NB cells. In this study, we further confirmed the role VEGF in NB through mouse xenograft model and clinical analysis from NB patients. In xenograft experiments, CRT overexpression effectively inhibited the tumor growth. In addition, the mRNA and protein levels of VEGF and differentiation marker GAP-43 were upregulated by induced CRT expression. However, no significant correlation between the expression level of VEGF and microvessel density was observed in human NB tumors, suggesting a novel mechanism of VEGF participating in NB tumorigenesis through an angiogenesis-independent pathway. In NB patients' samples, mRNA expression levels of CRT and VEGF were positively correlated. Furthermore, positive VEGF expression by immunostaining of NB tumors was found to correlate well with histological grade of differentiation and predicted a favorable prognosis. In conclusion, our findings suggest that VEGF is a favorable prognostic factor of NB and might affect NB tumor behavior through CRT-driven neuronal differentiation rather than angiogenesis that might shed light on a novel therapeutic strategy to improve the outcome of NB.


Subject(s)
Calreticulin/metabolism , Cell Differentiation , Gene Expression , Neuroblastoma/pathology , Neurons/physiology , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Disease Models, Animal , GAP-43 Protein/analysis , Heterografts , Humans , Immunohistochemistry , Mice , Neoplasm Transplantation , Neurons/drug effects , Prognosis
8.
Sci Rep ; 6: 27050, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27244685

ABSTRACT

Lysophosphatidic acid (LPA), a growth factor-like phospholipid, regulates numerous physiological functions, including cell proliferation and differentiation. In a previous study, we have demonstrated that LPA activates erythropoiesis by activating the LPA 3 receptor subtype (LPA3) under erythropoietin (EPO) induction. In the present study, we applied a pharmacological approach to further elucidate the functions of LPA receptors during red blood cell (RBC) differentiation. In K562 human erythroleukemia cells, knockdown of LPA2 enhanced erythropoiesis, whereas knockdown of LPA3 inhibited RBC differentiation. In CD34(+) human hematopoietic stem cells (hHSC) and K526 cells, the LPA3 agonist 1-oleoyl-2-methyl-sn-glycero-3-phosphothionate (2S-OMPT) promoted erythropoiesis, whereas the LPA2 agonist dodecyl monophosphate (DMP) and the nonlipid specific agonist GRI977143 (GRI) suppressed this process. In zebrafish embryos, hemoglobin expression was significantly increased by 2S-OMPT treatment but was inhibited by GRI. Furthermore, GRI treatment decreased, whereas 2S-OMPT treatment increased RBC counts and amount of hemoglobin level in adult BALB/c mice. These results indicate that LPA2 and LPA3 play opposing roles during RBC differentiation. The pharmacological activation of LPA receptor subtypes represent a novel strategies for augmenting or inhibiting erythropoiesis.


Subject(s)
Cell Differentiation/drug effects , Erythropoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Lysophospholipids/metabolism , Receptors, Lysophosphatidic Acid/genetics , Animals , Embryo, Nonmammalian , Erythrocytes/cytology , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythropoiesis/genetics , Erythropoietin/pharmacology , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hemoglobins/antagonists & inhibitors , Hemoglobins/biosynthesis , Hemoglobins/genetics , Humans , K562 Cells , Lysophospholipids/pharmacology , Mice , Mice, Inbred BALB C , Organophosphates/pharmacology , Organothiophosphates/pharmacology , Phosphatidic Acids/pharmacology , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/metabolism , Zebrafish
9.
Biochim Biophys Acta ; 1851(2): 172-83, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25463482

ABSTRACT

Erythrocytes and megakaryocytes (MK) are derived from a common progenitor that undergoes lineage specification. Lysophosphatidic acid (LPA), a lipid growth factor was previously shown to be a regulator for erythropoietic process through activating LPA receptor 3 (LPA3). However, whether LPA affects megakaryopoiesis remains unclear. In this study, we used K562 leukemia cell line as a model to investigate the roles of LPA in MK differentiation. We demonstrated that K562 cells express both LPA2 and LPA3, and the expression levels of LPA2 are higher than LPA3. Treatment with phorbol 12-myristate 13-acetate, a commonly used inducer of megakaryopoiesis, reciprocally regulates the expressions of LPA2 and LPA3. By pharmacological blockers and knockdown experiments, we showed that activation of LPA2 suppresses whereas, LPA3 promotes megakaryocytic differentiation in K562. The LPA2-mediated inhibition is dependent on ß-catenin translocation, whereas reactive oxygen species (ROS) generation is a downstream signal for activation of LPA3. Furthermore, the hematopoietic transcriptional factors GATA-1 and FLI-1, appear to be involved in these regulatory mechanisms. Taken together, our results suggested that LPA2 and LPA3 may function as a molecular switch and play opposing roles during megakaryopoiesis of K562 cells.


Subject(s)
Leukemia, Erythroblastic, Acute/metabolism , Megakaryocytes/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Thrombopoiesis , GATA1 Transcription Factor/metabolism , Humans , Integrin beta3/metabolism , K562 Cells , Leukemia, Erythroblastic, Acute/genetics , Megakaryocytes/drug effects , Microfilament Proteins/metabolism , RNA Interference , Reactive Oxygen Species/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Thrombopoiesis/drug effects , Time Factors , Trans-Activators , Transfection , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...