Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.407
Filter
1.
Front Immunol ; 13: 980911, 2022.
Article in English | MEDLINE | ID: mdl-36081495

ABSTRACT

Acute myelocytic leukemia (AML) is a malignancy of the stem cell precursors of the myeloid lineage. CD4+ and CD8+ T cells play pivotal roles in influencing AML progression but are functionally suppressed in the bone marrow microenvironment. We aimed to find hub genes related to T cell exhaustion and suppression, thereby providing evidence for immunotherapy. In this study, gene transcriptome expression data from TCGA and TARGET databases were utilized to find key genes. Firstly, CIBERSORT immune cell infiltration algorithm and WGCNA method were used to identify CD4+ and CD8+ T cells-related genes. Univariate and multivariate cox regression analyses were then introduced to construct the overall survival prognosis model and included hub genes. The ESTIMATE and ssGSEA scoring methods were used to analyze the correlation between the hub genes and immune activity. Single-cell transcriptome analysis was applied to detect the immune cells expressing hub genes, hence, to detect exact mechanisms. Consequently, FLT3LG and IFITM3P6 were determined to be positively correlated with patients' overall survival and microenvironment immune activity. Further study suggested FLT3-FLT3LG and IFITM3P6-miR-6748-3p-CBX7 signaling axes were involved in CD4+ and CD8+ T cells activation. This may be one of the mechanisms of T cells suppression in AML.


Subject(s)
Bone Marrow , CD8-Positive T-Lymphocytes , Hematopoietic Cell Growth Factors , Leukemia, Myeloid, Acute , Membrane Proteins , RNA-Binding Proteins , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Prognosis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Tumor Microenvironment
2.
Stem Cell Rev Rep ; 18(8): 3021-3032, 2022 12.
Article in English | MEDLINE | ID: mdl-35655001

ABSTRACT

Female germline stem cells (FGSCs) have been successfully isolated and characterized from postnatal mammalian and human ovarian tissues. However, the effects and mechanisms of action of natural small-molecule compounds on FGSCs are largely unknown. Here, we found that daidzein promoted the viability and proliferation of FGSCs. To elucidate the mechanism underlying this, we performed RNA-Sequence in daidzein-treated FGSCs and controls. The results showed that there were 153 upregulated and 156 downregulated genes in daidzein treatment. We confirmed the expression of some genes related to cell proliferation in the sequencing results by RT-PCR, such as Type C lectin domain family 11 member a (Clec11a), Mucin1 (Muc1), Glutathione peroxidase 3 (Gpx3), and Tet methylcytosine dioxygenase 1 (Tet1). The high expression of Clec11a at the protein level after daidzein treatment was also confirmed by western blotting. Furthermore, recombinant mouse Clec11a (rmClec11a) protein was shown to promote the viability and proliferation of FGSCs. However, knockdown of Clec11a inhibited the viability and proliferation of FGSCs, which could not be rescued by the administration of daidzein. These results indicate that daidzein promoted the viability and proliferation of FGSCs through Clec11a. In addition, both daidzein and rmClec11a activated the Akt signaling pathway in FGSCs. However, Clec11a knockdown inhibited this pathway, which could not be rescued by daidzein administration. Taken together, our findings revealed that daidzein activates the Akt signaling pathway to promote cell viability and proliferation through upregulating Clec11a. This study should deepen our understanding of the developmental mechanism of FGSCs and female infertility.


Subject(s)
Isoflavones , Oogonial Stem Cells , Animals , Female , Humans , Mice , Cell Proliferation , Isoflavones/pharmacology , Isoflavones/metabolism , Mammals/metabolism , Mixed Function Oxygenases/metabolism , Mixed Function Oxygenases/pharmacology , Oogonial Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Hematopoietic Cell Growth Factors/metabolism , Lectins, C-Type/metabolism , Up-Regulation
3.
J Natl Compr Canc Netw ; 20(5): 436-442, 2022 05.
Article in English | MEDLINE | ID: mdl-35545171

ABSTRACT

The NCCN Guidelines for Hematopoietic Growth Factors provide recommendations for the appropriate use of growth factors in the clinical management of febrile neutropenia (FN), chemotherapy-induced thrombocytopenia (CIT), and chemotherapy-induced anemia (CIA). Management and prevention of these sequelae are an integral part of supportive care for many patients undergoing cancer treatment. The purpose of these guidelines is to operationalize the evaluation, prevention, and treatment of FN, CIT, and CIA in adult patients with nonmyeloid malignancies and to enable the patient and clinician to assess management options for FN, CIT, and CIA in the context of an individual patient's condition. These NCCN Guidelines Insights provide a summary of the important recent updates to the NCCN Guidelines for Hematopoietic Growth Factors, with particular emphasis on the incorporation of a newly developed section on CIT.


Subject(s)
Anemia , Antineoplastic Agents , Neoplasms , Adult , Anemia/chemically induced , Anemia/drug therapy , Antineoplastic Agents/adverse effects , Hematopoietic Cell Growth Factors/therapeutic use , Humans , Neoplasms/drug therapy
4.
Signal Transduct Target Ther ; 6(1): 418, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34893580

ABSTRACT

The systemic processes involved in the manifestation of life-threatening COVID-19 and in disease recovery are still incompletely understood, despite investigations focusing on the dysregulation of immune responses after SARS-CoV-2 infection. To define hallmarks of severe COVID-19 in acute disease (n = 58) and in disease recovery in convalescent patients (n = 28) from Hannover Medical School, we used flow cytometry and proteomics data with unsupervised clustering analyses. In our observational study, we combined analyses of immune cells and cytokine/chemokine networks with endothelial activation and injury. ICU patients displayed an altered immune signature with prolonged lymphopenia but the expansion of granulocytes and plasmablasts along with activated and terminally differentiated T and NK cells and high levels of SARS-CoV-2-specific antibodies. The core signature of seven plasma proteins revealed a highly inflammatory microenvironment in addition to endothelial injury in severe COVID-19. Changes within this signature were associated with either disease progression or recovery. In summary, our data suggest that besides a strong inflammatory response, severe COVID-19 is driven by endothelial activation and barrier disruption, whereby recovery depends on the regeneration of the endothelial integrity.


Subject(s)
Antibodies, Viral/blood , Blood Proteins/metabolism , COVID-19/diagnosis , Cytokine Release Syndrome/diagnosis , Endothelium, Vascular/virology , Lymphopenia/diagnosis , SARS-CoV-2/pathogenicity , Biomarkers/blood , C-Reactive Protein/metabolism , COVID-19/immunology , COVID-19/mortality , COVID-19/virology , Chemokine CXCL10/blood , Chemokine CXCL9/blood , Cluster Analysis , Convalescence , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/mortality , Cytokine Release Syndrome/virology , Disease Progression , Endothelium, Vascular/immunology , Granulocytes/immunology , Granulocytes/virology , Hematopoietic Cell Growth Factors/blood , Hepatocyte Growth Factor/blood , Humans , Intensive Care Units , Interleukin-12 Subunit p40/blood , Interleukin-6/blood , Interleukin-8/blood , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Lectins, C-Type/blood , Lymphopenia/immunology , Lymphopenia/mortality , Lymphopenia/virology , Plasma Cells/immunology , Plasma Cells/virology , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/virology
5.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Article in English | MEDLINE | ID: mdl-34140410

ABSTRACT

We previously described a new osteogenic growth factor, osteolectin/Clec11a, which is required for the maintenance of skeletal bone mass during adulthood. Osteolectin binds to Integrin α11 (Itga11), promoting Wnt pathway activation and osteogenic differentiation by leptin receptor+ (LepR+) stromal cells in the bone marrow. Parathyroid hormone (PTH) and sclerostin inhibitor (SOSTi) are bone anabolic agents that are administered to patients with osteoporosis. Here we tested whether osteolectin mediates the effects of PTH or SOSTi on bone formation. We discovered that PTH promoted Osteolectin expression by bone marrow stromal cells within hours of administration and that PTH treatment increased serum osteolectin levels in mice and humans. Osteolectin deficiency in mice attenuated Wnt pathway activation by PTH in bone marrow stromal cells and reduced the osteogenic response to PTH in vitro and in vivo. In contrast, SOSTi did not affect serum osteolectin levels and osteolectin was not required for SOSTi-induced bone formation. Combined administration of osteolectin and PTH, but not osteolectin and SOSTi, additively increased bone volume. PTH thus promotes osteolectin expression and osteolectin mediates part of the effect of PTH on bone formation.


Subject(s)
Hematopoietic Cell Growth Factors/metabolism , Lectins, C-Type/metabolism , Osteogenesis/drug effects , Parathyroid Hormone/pharmacology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cancellous Bone/drug effects , Cancellous Bone/pathology , Female , Hematopoietic Cell Growth Factors/blood , Hematopoietic Cell Growth Factors/deficiency , Humans , Lectins, C-Type/blood , Lectins, C-Type/deficiency , Mice, Inbred C57BL , Organ Size/drug effects , Osteoporosis/blood , Premenopause/blood , Wnt Signaling Pathway/drug effects
6.
PLoS Negl Trop Dis ; 15(3): e0008906, 2021 03.
Article in English | MEDLINE | ID: mdl-33755669

ABSTRACT

Chagas' disease (CD), caused by the hemoflagellate protozoan, Trypanosoma cruzi, is endemic in most countries of Latin America. Heart failure (HF) is often a late manifestation of chronic CD, and is associated with high morbidity and mortality. Inflammatory processes mediated by cytokines play a key role in the pathogenesis and progression of CD. Keeping in view the inflammatory nature of CD, this study investigated the possible role of 21 different inflammatory cytokines as biomarkers for prediction and prognosis of CD. The plasma concentration of these cytokines was measured in a group of patients with CD (n = 94), and then compared with those measured in patients with dilated cardiomyopathy (DCM) from idiopathic causes (n = 48), and with control subjects (n = 25). Monovariately, plasma levels of cytokines such as stem cell growth factor beta (SCGF beta), hepatocyte growth factor (HGF), monokine induced by interferon gamma (CXCL9), and macrophage inhibitory factor (MIF) were significantly increased in CD patients with advanced HF compared to control group. None of the cytokines could demonstrate any prognostic potency in CD patients, and only MIF and stromal derived factor-1 alpha (CXCL12) showed significance in predicting mortality and necessity for heart transplant in DCM patients. However, multivariate analysis prognosticated a large proportion of CD and DCM patients. In CD patients, HGF and Interleukin-12p40 (IL-12p40) together separated 81.9% of 3-year survivors from the deceased, while in DCM patients, CXCL12, stem cell factor (SCF), and CXCL9 together discriminated 77.1% of survivors from the deceased. The significant increase in plasma concentrations of cytokines such as HGF and CXCL9 in CD patients, and the ability of these cytokines to prognosticate a large proportion of CD and DCM patients multivariately, encourages further studies to clarify the diagnostic and prognostic potential of cytokines in such patients.


Subject(s)
Cardiomyopathy, Dilated/blood , Cardiomyopathy, Dilated/mortality , Chagas Disease/diagnosis , Chagas Disease/mortality , Cytokines/blood , Biomarkers/blood , Chagas Disease/blood , Chagas Disease/pathology , Chemokine CXCL9/blood , Female , Heart Failure/mortality , Heart Failure/parasitology , Hematopoietic Cell Growth Factors/blood , Hepatocyte Growth Factor/blood , Humans , Intramolecular Oxidoreductases/blood , Lectins, C-Type/blood , Macrophage Migration-Inhibitory Factors/blood , Male , Middle Aged , Prognosis , Trypanosoma cruzi/physiology
7.
Am J Obstet Gynecol ; 225(1): 65.e1-65.e14, 2021 07.
Article in English | MEDLINE | ID: mdl-33539826

ABSTRACT

BACKGROUND: Ovarian senescence is a normal age-associated phenomenon, but increasingly younger women are affected by diminished ovarian reserves or premature ovarian insufficiency. There is an urgent need for developing therapies to improve ovarian function in these patients. In this context, previous studies suggest that stem cell-secreted factors could have regenerative properties in the ovaries. OBJECTIVE: This study aimed to test the ability of various human plasma sources, enriched in stem cell-secreted factors, and the mechanisms behind their regenerative properties, to repair ovarian damage and to promote follicular development. STUDY DESIGN: In the first phase, the effects of human plasma enriched in bone marrow stem cell soluble factors by granulocyte colony-stimulating factor mobilization, umbilical cord blood plasma, and their activated forms on ovarian niche, follicle development, and breeding performance were assessed in mouse models of chemotherapy-induced ovarian damage (n=7 per group). In addition, the proteomic profile of each plasma was analyzed to find putative proteins and mechanism involved in their regenerative properties in ovarian tissue. In the second phase, the most effective plasma treatment was validated in human ovarian cortex xenografted in immunodeficient mice (n=4 per group). RESULTS: Infusion of human plasma enriched bone marrow stem cell soluble factors by granulocyte colony-stimulating factor mobilization or of umbilical cord blood plasma-induced varying degrees of microvessel formation and cell proliferation and reduced apoptosis in ovarian tissue to rescue follicular development and fertility in mouse models of ovarian damage. Plasma activation enhanced these effects. Activated granulocyte colony-stimulating factor plasma was the most potent inducing ovarian rescue in both mice and human ovaries, and proteomic analysis indicated that its effects may be mediated by soluble factors related to cell cycle and apoptosis, gene expression, signal transduction, cell communication, response to stress, and DNA repair of double-strand breaks, the most common form of age-induced damage in oocytes. CONCLUSION: Our findings suggested that stem cell-secreted factors present in both granulocyte colony-stimulating factor-mobilized and umbilical cord blood plasma could be an effective treatment for increasing the reproductive outcomes in women with impaired ovarian function owing to several causes. The activated granulocyte colony-stimulating factor plasma, which is already enriched in both stem cell-secreted factors and platelet-enclosed growth factors, seems to be the most promising treatment because of its most potent restorative effects on the ovary together with the autologous source.


Subject(s)
Hematopoietic Cell Growth Factors/therapeutic use , Ovarian Follicle/drug effects , Ovarian Reserve/drug effects , Ovary/drug effects , Primary Ovarian Insufficiency/drug therapy , Stem Cells/metabolism , Animals , Bone Marrow Cells , Disease Models, Animal , Female , Fetal Blood , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Heterografts , Humans , Infant, Newborn , Mice , Mice, Inbred NOD , Ovarian Follicle/growth & development , Ovary/transplantation , Plasma/chemistry , Stem Cell Factor/pharmacology
8.
Nature ; 591(7850): 438-444, 2021 03.
Article in English | MEDLINE | ID: mdl-33627868

ABSTRACT

Stromal cells in adult bone marrow that express leptin receptor (LEPR) are a critical source of growth factors, including stem cell factor (SCF), for the maintenance of haematopoietic stem cells and early restricted progenitors1-6. LEPR+ cells are heterogeneous, including skeletal stem cells and osteogenic and adipogenic progenitors7-12, although few markers have been available to distinguish these subsets or to compare their functions. Here we show that expression of an osteogenic growth factor, osteolectin13,14, distinguishes peri-arteriolar LEPR+ cells poised to undergo osteogenesis from peri-sinusoidal LEPR+ cells poised to undergo adipogenesis (but retaining osteogenic potential). Peri-arteriolar LEPR+osteolectin+ cells are rapidly dividing, short-lived osteogenic progenitors that increase in number after fracture and are depleted during ageing. Deletion of Scf from adult osteolectin+ cells did not affect the maintenance of haematopoietic stem cells or most restricted progenitors but depleted common lymphoid progenitors, impairing lymphopoiesis, bacterial clearance, and survival after acute bacterial infection. Peri-arteriolar osteolectin+ cell maintenance required mechanical stimulation. Voluntary running increased, whereas hindlimb unloading decreased, the frequencies of peri-arteriolar osteolectin+ cells and common lymphoid progenitors. Deletion of the mechanosensitive ion channel PIEZO1 from osteolectin+ cells depleted osteolectin+ cells and common lymphoid progenitors. These results show that a peri-arteriolar niche for osteogenesis and lymphopoiesis in bone marrow is maintained by mechanical stimulation and depleted during ageing.


Subject(s)
Arterioles , Lymphopoiesis , Osteogenesis , Stem Cell Niche , Adipose Tissue/cytology , Aging , Animals , Bone Marrow Cells/cytology , Bone and Bones/cytology , Female , Hematopoietic Cell Growth Factors/metabolism , Lectins, C-Type/metabolism , Lymphocytes/cytology , Male , Mice , Receptors, Leptin/metabolism , Stem Cell Factor , Stromal Cells/cytology
9.
Blood Rev ; 45: 100690, 2021 01.
Article in English | MEDLINE | ID: mdl-32273121

ABSTRACT

Exposure to acute, high-dose, high dose-rate whole-body ionizing radiations damages the bone marrow resulting in rapid decreases in concentrations of blood cells, especially lymphocytes, granulocytes and platelets with associated risks of infection and bleeding. In several experimental models including non-human primate radiation exposure models giving molecularly cloned haematopoietic growth factor including granulocyte/macrophage colony-stimulating factor (G/M-CSF; sargramostim) and granulocyte colony-stimulating factor (G-CSF; filgrastim and pegylated G-CSF [peg-filgrastim]) accelerates bone marrow recovery and increases survival. Based on these data these molecules are US FDA approved for treating victims of radiation and nuclear incidents, accident and events such as nuclear terrorism and are included in the US National Strategic Stockpile. We discuss the immediate medical response to these events including how to estimate radiation dose and uniformity and which interventions are appropriate in different radiation exposures settings. We also discuss similarities and differences between molecularly cloned haematopoietic growth factors.


Subject(s)
Hematopoietic Cell Growth Factors/administration & dosage , Radiation Dosage , Radiation, Ionizing , Recombinant Proteins/administration & dosage , Whole-Body Irradiation/adverse effects , Bone Marrow/drug effects , Bone Marrow/radiation effects , Disease Management , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Humans , Radiation Injuries/drug therapy , Radiation Injuries/etiology
10.
Cell Rep ; 33(2): 108252, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053358

ABSTRACT

Osteogenic suppressors such as Sclerostin not only regulate skeletal development and regeneration but also serve as anti-osteoporosis drug targets. However, very few druggable suppressors have been identified due to limited understanding of the molecular mechanisms governing osteogenesis. Here, we show that fibroblast activation protein (Fap), a serine protease inhibited by the bone growth factor Osteolectin, is an osteogenic suppressor. Genetic deletion of Fap significantly ameliorates limb trabecular bone loss during aging. Pharmacological inhibition of Fap significantly promotes bone formation and inhibits bone resorption in wild-type mice by differentially regulating canonical Wnt and nuclear factor κB (NF-κB) pathways. Pharmacological inhibition of Fap promotes osteoblast differentiation, inhibits osteoclast differentiation, and significantly attenuates osteoporosis in ovariectomized mice. Epistasis analyses in zebrafish show that Osteolectin functions as an endogenous inhibitor of Fap to promote vertebrae mineralization. Taken together, we identify Fap as an important osteogenic suppressor and a potential drug target to treat osteoporosis.


Subject(s)
Endopeptidases/metabolism , Membrane Proteins/metabolism , Molecular Targeted Therapy , Osteogenesis , Osteoporosis/drug therapy , Aging/metabolism , Aging/pathology , Animals , Bone Resorption/complications , Bone Resorption/diagnostic imaging , Bone Resorption/pathology , Calcification, Physiologic , Cell Differentiation , Epistasis, Genetic , Gene Deletion , HEK293 Cells , Hematopoietic Cell Growth Factors/metabolism , Humans , Lectins, C-Type/metabolism , Membrane Proteins/antagonists & inhibitors , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Osteoporosis/complications , Osteoporosis/diagnostic imaging , Osteoporosis/pathology , Ovariectomy , Peptide Hydrolases/metabolism , Protein Binding , Zebrafish , Zebrafish Proteins/metabolism
11.
Exp Hematol ; 91: 39-45.e2, 2020 11.
Article in English | MEDLINE | ID: mdl-32961298

ABSTRACT

Mutations in the gene encoding DNA methyltransferase 3A (DNMT3A) comprise the majority of mutations found in clonal hematopoiesis (CH), an age-related condition that was recently found to affect outcomes in patients undergoing hematopoietic stem cell transplant (HSCT). Recent studies have indicated that patients with CH have worse prognoses after HSCT, suggesting stress imposed by HSCT preconditioning agents may impact hematopoietic stem cell (HSC) dynamics in transplant recipients. In this study, we used a competitive transplantation mouse model to investigate how treatment with the common preconditioning agents 5-fluorouracil (5-FU) and busulfan (BU) affect the prevalence of Dnmt3a-/- HSCs and progenitor cells in competition with wild-type cells. We found that, though sufficient to deplete peripheral blood counts, 5-FU preconditioning did not significantly alter the frequency of Dnmt3a-null hematopoietic stem and progenitor cells (HSPCs) in mosaic mice. In contrast, mice treated with BU had a sevenfold decline in total bone marrow cells and an increase in Dnmt3a-null HSPCs that was detectable in peripheral blood. Indeed, even though all mosaic mice had a starting engraftment of ∼10%-40%, 85%-100% of HSPCs were Dnmt3a-null in four of seven mice after BU treatment, indicating these cells expand dramatically during recovery. Overall, these results suggest that individual preconditioning regimens have different effects on the expansion of Dnmt3a-mutant cells in patients with pre-existing CH. Thus, the presence of CH-associated mutants should be evaluated prior to selecting preconditioning regimens for HSCT.


Subject(s)
Busulfan/pharmacology , DNA (Cytosine-5-)-Methyltransferases/deficiency , Hematopoiesis/genetics , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Animals , Bone Marrow/pathology , Cell Division/drug effects , Cell Lineage , Clone Cells , DNA Methyltransferase 3A , Fluorouracil/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Mice , Radiation Chimera
12.
J Cell Physiol ; 235(10): 6357-6365, 2020 10.
Article in English | MEDLINE | ID: mdl-32003015

ABSTRACT

C-type lectin domain family 11 member A (Clec11a), also known as stem cell growth factor (SCGF), C-type lectin superfamily member 3 (CLECSF3), or osteolectin was initially identified as a growth factor for hematopoietic progenitor cells. The human Clec11a gene encodes a polypeptide of 323 amino acids with characteristics of a secreted glycoprotein encompassing two integrin-binding motifs, RGD (Arg-Gly-Asp) and LDT (Leu-Asp-Thr), a putative leucine zipper domain, and a functional C-type lectin domain. It regulates hematopoietic differentiation and homeostasis and exhibits a protective effect against severe malarial anemia and lipotoxicity. Furthermore, Clec11a promotes the differentiation of mesenchymal progenitors into mature osteoblasts in vitro and plays an important role in the maintenance of adult skeleton age-related bone loss and fracture repair. Receptor ligand binding results in activation of downstream signaling cascades including glycogen synthase kinase 3 (GSK3), ß-catenin, and Wnt, resulting in the expression of osteoblast-related gene transcripts including Alp, Runx2, Lef1, and Axin2. In addition, Clec11a is also associated with the development of several cancers, including leukemia, multiple myeloma, and gastrointestinal tract tumors. To date, however, the mechanisms governing transcription regulation of the Clec11a gene are not known and remain to be uncovered. Understanding the function and mechanism of action of Clec11a will pave the way for the development of Clec11a as a novel therapeutic target for conditions such as cancer, anemia, and skeletal diseases.


Subject(s)
Hematopoietic Cell Growth Factors/genetics , Neoplasms/genetics , Amino Acid Sequence , Animals , Biology , Humans , Molecular Structure , Transcription, Genetic/genetics
13.
Nat Commun ; 11(1): 666, 2020 02 03.
Article in English | MEDLINE | ID: mdl-32015345

ABSTRACT

Inflammatory signals arising from the microenvironment have emerged as critical regulators of hematopoietic stem cell (HSC) function during diverse processes including embryonic development, infectious diseases, and myelosuppressive injuries caused by irradiation and chemotherapy. However, the contributions of cellular subsets within the microenvironment that elicit niche-driven inflammation remain poorly understood. Here, we identify endothelial cells as a crucial component in driving bone marrow (BM) inflammation and HSC dysfunction observed following myelosuppression. We demonstrate that sustained activation of endothelial MAPK causes NF-κB-dependent inflammatory stress response within the BM, leading to significant HSC dysfunction including loss of engraftment ability and a myeloid-biased output. These phenotypes are resolved upon inhibition of endothelial NF-κB signaling. We identify SCGF as a niche-derived factor that suppresses BM inflammation and enhances hematopoietic recovery following myelosuppression. Our findings demonstrate that chronic endothelial inflammation adversely impacts niche activity and HSC function which is reversible upon suppression of inflammation.


Subject(s)
Endothelial Cells/metabolism , Hematopoiesis/physiology , Hematopoietic Cell Growth Factors/metabolism , Lectins, C-Type/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Animals , Antigens, CD , Bone Marrow , Cadherins , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Inflammation , Male , Mice , Signal Transduction , Transplantation, Autologous
14.
Theranostics ; 10(5): 2293-2308, 2020.
Article in English | MEDLINE | ID: mdl-32089743

ABSTRACT

Osteoporosis and osteoporotic fractures severely compromise quality of life in elderly people and lead to early death. Human umbilical cord mesenchymal stromal cell (MSC)-derived extracellular vesicles (hucMSC-EVs) possess considerable therapeutic effects in tissue repair and regeneration. Thus, in the present study, we investigated the effects of hucMSC-EVs on primary and secondary osteoporosis and explored the underlying mechanisms. Methods: hucMSCs were isolated and cultured. EVs were obtained from the conditioned medium of hucMSCs and determined by using transmission electron microscopy, dynamic light scattering and Western Blot analyses. The effects of hucMSC-EVs on ovariectomy-induced postmenopausal osteoporosis and tail suspension-induced hindlimb disuse osteoporosis in mouse models were assessed by using microcomputed tomography, biomechanical, histochemical and immunohistochemical, as well as histomorphometric analyses. Proteomic analysis was applied between hucMSC-EVs and hucMSCs to screen the candidate proteins that mediate hucMSC-EVs function. The effects of hucMSC-EVs on osteogenic and adipogenic differentiation of bone marrow mesenchymal stromal cells (BMSCs), and osteoclastogenesis of the macrophage cell line RAW264.7 in vitro were determined by using cytochemical staining and quantitative real-time PCR analysis. Subsequently, the roles of the key protein in hucMSC-EVs-induced regulation on BMSCs and RAW264.7 cells were evaluated. Results: hucMSCs were able to differentiate into osteoblasts, adipocytes or chondrocytes and positively expressed CD29, CD44, CD73 and CD90, but negatively expressed CD34 and CD45. The morphological assessment revealed the typical cup- or sphere-shaped morphology of hucMSC-EVs with diameters predominantly ranging from 60 nm to 150 nm and expressed CD9, CD63, CD81 and TSG101. The systemic administration of hucMSC-EVs prevented bone loss and maintained bone strength in osteoporotic mice by enhancing bone formation, reducing marrow fat accumulation and decreasing bone resorption. Proteomic analysis showed that the potently pro-osteogenic protein, CLEC11A (C-type lectin domain family 11, member A) was very highly enriched in hucMSC-EVs. In addition, hucMSC-EVs enhanced the shift from adipogenic to osteogenic differentiation of BMSCs via delivering CLEC11A in vitro. Moreover, CLEC11A was required for the inhibitory effects of hucMSC-EVs on osteoclast formation. Conclusion: Our results suggest that hucMSC-EVs serve as a critical regulator of bone metabolism by transferring CLEC11A and may represent a potential agent for prevention and treatment of osteoporosis.


Subject(s)
Bone and Bones/metabolism , Extracellular Vesicles/metabolism , Hematopoietic Cell Growth Factors/metabolism , Lectins, C-Type/metabolism , Mesenchymal Stem Cells/metabolism , Osteoporosis/metabolism , Umbilical Cord/metabolism , Adipocytes/metabolism , Adipogenesis , Animals , Bone Marrow/metabolism , Cell Differentiation , Chondrocytes/metabolism , Disease Models, Animal , Humans , Mice , Osteoblasts/metabolism , Osteogenesis , Osteoporosis/pathology , Proteomics , RAW 264.7 Cells , Umbilical Cord/cytology , X-Ray Microtomography
15.
J Natl Compr Canc Netw ; 18(1): 12-22, 2020 01.
Article in English | MEDLINE | ID: mdl-31910384

ABSTRACT

Management of febrile neutropenia (FN) is an integral part of supportive care for patients undergoing cancer treatment. The NCCN Guidelines for Hematopoietic Growth Factors provide suggestions for appropriate evaluation, risk determination, prophylaxis, and management of FN. These NCCN Guidelines are intended to guide clinicians in the appropriate use of growth factors for select patients undergoing treatment of nonmyeloid malignancies. These NCCN Guidelines Insights highlight important updates to the NCCN Guidelines regarding the incorporation of newly FDA-approved granulocyte-colony stimulating factor biosimilars for the prevention and treatment of FN.


Subject(s)
Biosimilar Pharmaceuticals/therapeutic use , Chemotherapy-Induced Febrile Neutropenia/drug therapy , Hematopoietic Cell Growth Factors/therapeutic use , Neoplasms/drug therapy , Practice Guidelines as Topic , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biosimilar Pharmaceuticals/economics , Biosimilar Pharmaceuticals/standards , Chemotherapy-Induced Febrile Neutropenia/etiology , Drug Approval , Drug Costs , Education, Medical, Continuing , Hematopoietic Cell Growth Factors/economics , Hematopoietic Cell Growth Factors/standards , Humans , Medical Oncology/education , Medical Oncology/standards , Neoplasms/blood , Oncologists/education , Organizations, Nonprofit/standards , Risk Factors , United States , United States Food and Drug Administration/legislation & jurisprudence
16.
Article in English | MEDLINE | ID: mdl-31988205

ABSTRACT

The hematopoietic system is highly organized to maintain its functional integrity and to meet lifelong organismal demands. Hematopoietic stem cells (HSCs) must balance self-renewal with differentiation and the regeneration of the blood system. It is a complex balancing act between these competing HSC functions. Although highly quiescent at steady state, HSCs become activated in response to inflammatory cytokines and regenerative challenges. This activation phase leads to many intrinsic stresses such as replicative, metabolic, and oxidative stress, which can cause functional decline, impaired self-renewal, and exhaustion of HSCs. To cope with these insults, HSCs use both built-in and emergency-triggered stress-response mechanisms to maintain homeostasis and to defend against disease development. In this review, we discuss how the hematopoietic system operates in steady state and stress conditions, what strategies are used to maintain functional integrity, and how deregulation in the balance between self-renewal and regeneration can drive malignant transformation.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Regeneration/physiology , Animals , Antineoplastic Agents, Immunological , Cell Differentiation , Flow Cytometry/methods , Hematopoietic Cell Growth Factors/blood , Hematopoietic Stem Cells/cytology , Homeostasis , Humans , Mice
17.
Handb Exp Pharmacol ; 261: 257-283, 2020.
Article in English | MEDLINE | ID: mdl-31451971

ABSTRACT

This review focuses on certain hematopoietic growth factors that are used as medications in clinical neonatology. It is important to note at the chapter onset that although all of the pharmacological agents mentioned in this review have been approved by the US Food and Drug administration for use in humans, none have been granted a specific FDA indication for neonates. Thus, in a sense, all of the agents mentioned in this chapter could be considered experimental, when used in neonates. However, a great many of the pharmacological agents utilized routinely in neonatology practice do not have a specific FDA indication for this population of patients. Consequently, many of the agents reviewed in this chapter are considered by some practitioners to be nonexperimental and are used when they judge such use to be "best practice" for the disorders under treatment.The medicinal uses of the agents in this chapter vary considerably, between geographic locations, and sometimes even within an institutions. "Consistent approaches" aimed at using these agents in uniform ways in the practice of neonatology are encouraged. Indeed some healthcare systems, and some individual NICUs, have developed written guidelines for using these agents within the practice group. Some such guidelines are provided in this review. It should be noted that these guidelines, or "consistent approaches," must be viewed as dynamic and changing, requiring adjustment and refinement as additional evidence accrues.


Subject(s)
Hematopoietic Cell Growth Factors/chemistry , Neonatology , Hematopoietic Cell Growth Factors/metabolism , Humans , Infant, Newborn
18.
Bull Cancer ; 107(1S): S44-S51, 2020 Jan.
Article in French | MEDLINE | ID: mdl-31570213

ABSTRACT

The modalities of mobilization of hematopoietic stem cells in autologous transplantation have evolved in recent years. The Francophone Society of Bone Marrow Transplantation and Cellular Therapy (SFGM-TC) organized the 9th hematopoietic stem cell transplantation clinical practices harmonization workshop series in September 2018 in Lille, France, to conduct a review of current practices of the society centers and of international recommendations. The cell dose objectives have been revised. The modalities of mobilization including the use of plerixafor have been specified allowing reaching the objectives of collection while limiting the number of apheresis. Collections failures have become exceptional.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation , Algorithms , Antigens, CD34/analysis , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Benzylamines , Blood Component Removal/methods , Bone Marrow/drug effects , Cell Count , Cell Separation/methods , Cyclams , Granulocyte Colony-Stimulating Factor/adverse effects , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cell Mobilization/standards , Heterocyclic Compounds/pharmacology , Humans , Practice Patterns, Physicians' , Risk Factors , Transplantation, Autologous
19.
Exp Cell Res ; 384(1): 111613, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31494095

ABSTRACT

The lipotoxicity is considered as one of the risk for diabetes. Here we report C-type lectin domain family 11, member A (Clec11a) as a new regulator in islet playing a protective role in lipotoxicity induced dysfunction. Islet transcriptome sequencing was performed using the high-fat diet induced obesity (DIO) mice model. We found a significant decrease of Clec11a expression in islets of DIO mice compared to normal control mice, which was further confirmed by real-time PCR. Immunostaining demonstrated the localization of the Clec11a protein in mouse islets. Administration of recombinant human Clec11a (rClec11a) protein promoted the proliferation of islet cells and rescued the inhibition of fatty acid on cell proliferation, which involved the activation of Erk signaling pathway. We also found that the rClec11a altered the expression of genes involved in lipid metabolism.


Subject(s)
Cell Proliferation/physiology , Hematopoietic Cell Growth Factors/metabolism , Islets of Langerhans/metabolism , Lectins, C-Type/metabolism , Lipid Metabolism/physiology , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Signal Transduction/physiology , Transcriptome/physiology
20.
PLoS Biol ; 17(7): e3000350, 2019 07.
Article in English | MEDLINE | ID: mdl-31265461

ABSTRACT

Mutagenic screening is powerful for identifying key genes involved in developmental processes. However, such screens are successful only in lower organisms. Here, we develop a targeted genetic screening approach in mice through combining androgenetic haploid embryonic stem cells (AG-haESCs) and clustered regularly interspaced palindromic repeats/CRISPR-associated protein 9 (CRISPR-Cas9) technology. We produced a mutant semi-cloned (SC) mice pool by oocyte injection of AG-haESCs carrying constitutively expressed Cas9 and an single guide RNA (sgRNA) library targeting 72 preselected genes in one step and screened for bone-development-related genes through skeletal analysis at birth. This yielded 4 genes: Zic1 and Clec11a, which are required for bone development, and Rln1 and Irx5, which had not been previously considered. Whereas Rln1-/- mice exhibited small skeletal size only at birth, Irx5-/- mice showed skeletal abnormalities both in postnatal and adult phases due to decreased bone mass and increased bone marrow adipogenesis. Mechanistically, iroquois homeobox 5 (IRX5) promotes osteoblastogenesis and inhibits adipogenesis by suppressing peroxisome proliferator activated receptor γ (PPARγ) activation. Thus, AG-haESC-mediated functional mutagenic screening opens new avenues for genetic interrogation of developmental processes in mice.


Subject(s)
Bone Development/genetics , Gene Expression Regulation, Developmental , Gene Targeting/methods , Genetic Testing/methods , Mouse Embryonic Stem Cells/metabolism , Animals , CRISPR-Cas Systems , Cells, Cultured , Haploidy , Hematopoietic Cell Growth Factors/genetics , Hematopoietic Cell Growth Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice , Mice, Knockout , Relaxin/genetics , Relaxin/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL