Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 413
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167025, 2024 03.
Article En | MEDLINE | ID: mdl-38237741

BACKGROUND: Bone marrow mesenchymal stem cells (BM-MSC) are an integral part of the BM niche that is essential to maintain hematopoietic homeostasis. In aplastic anemia (AA), a few studies have reported phenotypic defects in the BM-MSC, such as reduced proliferation, imbalanced differentiation, and apoptosis; however, the alterations at the molecular level need to be better characterized. Therefore, the current study aims to identify the causative factors underlying the compromised functions of AA BM-MSC that might eventually be contributing to the AA pathobiology. METHODS: We performed RNA sequencing (RNA-Seq) using the Illumina platform to comprehend the distinction between the transcriptional landscape of AA and control BM-MSC. Further, we validated the alterations observed in senescence by Senescence- associated beta-galactosidase (SA -ß-gal) assay, DNA damage by γH2AX staining, and telomere attrition by relative telomere length assessment and telomerase activity assay. We used qRT-PCR to analyze changes in some of the genes associated with these molecular mechanisms. RESULTS: The transcriptome profiling revealed enrichment of senescence-associated genes and pathways in AA BM-MSC. The senescent phenotype of AA BM-MSC was accompanied by enhanced SA -ß-gal activity and elevated expression of senescence associated genes TP53, PARP1, and CDKN1A. Further, we observed increased γH2AX foci indicating DNA damage, reduced telomere length, and diminished telomerase activity in the AA BM-MSC. CONCLUSION: Our results highlight that AA BM-MSC have a senescent phenotype accompanied by other cellular defects like DNA damage and telomere attrition, which are most likely driving the senescent phenotype of AA BM-MSC thus hampering their hematopoiesis supporting properties as observed in AA.


Anemia, Aplastic , Mesenchymal Stem Cells , Telomerase , Humans , Anemia, Aplastic/genetics , Anemia, Aplastic/metabolism , Telomerase/genetics , Telomerase/metabolism , Mesenchymal Stem Cells/metabolism , Telomere/genetics , DNA Repair
2.
Tissue Cell ; 86: 102287, 2024 Feb.
Article En | MEDLINE | ID: mdl-38086146

BACKGROUND: Increasing evidence indicate that enhanced adipogenic differentiation of bone marrow mesenchymal stem cells (BM-MSCs) could contribute to the adiposity alteration in marrow microenvironment of aplastic anemia (AA). Identifying small molecule drugs with role in inhibiting adipogenesis of BM-MSCs may represent a novel direction in AA therapy by improving BM-MSCs mediated marrow microenvironment. METHODS: For the purpose, we isolated AA BM-MSCs through whole bone marrow cell culture, evaluated a series of small molecule drugs using the in vitro adipogenic differentiation model of BM-MSCs, and finally focused on emodin, a natural anthraquinone derivative. Subsequently, we systematically investigated the molecular mechanism of emodin in attenuating adipogenic process by means of microarray profiling, bioinformatics analysis and lentivirus-mediated functional studies and rescue assay. RESULTS: We found that emodin presented significantly suppressive effect on the in vitro adipogenic differentiation of AA BM-MSCs. Further mechanistic investigation revealed that emodin could increase the expression of Tribbles homolog 3 (TRIB3) which exhibited remarkably decreased expression in AA BM-MSCs compared with the normal counterparts and was subsequently demonstrated as a negative regulator in adipogenesis of AA BM-MSCs. Besides, TRIB3 depletion alleviated the suppressive effect of emodin on the adipogenic differentiation of AA BM-MSCs. CONCLUSION: Our findings propose that emodin mediated TRIB3 up-regulation alleviates the adipogenic capacity of AA BM-MSCs, and emodin could serve as a potential therapeutic regimen for AA therapy.


Anemia, Aplastic , Emodin , Mesenchymal Stem Cells , Humans , Adipogenesis/genetics , Anemia, Aplastic/drug therapy , Anemia, Aplastic/metabolism , Bone Marrow , Emodin/pharmacology , Bone Marrow Cells , Cell Differentiation , Cells, Cultured , Repressor Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/metabolism
3.
Metabolomics ; 19(12): 94, 2023 Nov 17.
Article En | MEDLINE | ID: mdl-37975930

INTRODUCTION: Idiopathic acquired aplastic anemia (AA) is a bone marrow failure disorder where aberrant T-cell functions lead to depletion of hematopoietic stem and progenitor cells in the bone marrow (BM) microenvironment. T-cells undergo metabolic rewiring, which regulates their proliferation and differentiation. Therefore, studying metabolic variation in AA patients may aid us with a better understanding of the T-cell regulatory pathways governed by metabolites and their pathological engagement in the disease. OBJECTIVE: To identify the differential metabolites in BM plasma of AA patients, AA follow-up (AAF) in comparison to normal controls (NC) and to identify potential disease biomarker(s). METHODS: The study used 1D 1H NMR Carr-Purcell-Meiboom-Gill (CPMG) spectra to identify the metabolites present in the BM plasma samples of AA (n = 40), AAF (n = 16), and NC (n = 20). Metabolic differences between the groups and predictive biomarkers were identified by using multivariate analysis and receiver operating characteristic (ROC) module of Metaboanalyst V5.0 tool, respectively. RESULTS: The AA and AAF samples were well discriminated from NC group as per Principal Component analysis (PCA). Further, we found significant alteration in the levels of 17 metabolites in AA involved in amino-acid (Leucine, serine, threonine, phenylalanine, lysine, histidine, valine, tyrosine, and proline), carbohydrate (Glucose, lactate and mannose), fatty acid (Acetate, glycerol myo-inositol and citrate), and purine metabolism (hypoxanthine) in comparison to NC. Additionally, biomarker analysis predicted Hypoxanthine and Acetate can be used as a potential biomarker. CONCLUSION: The study highlights the significant metabolic alterations in the BM plasma of AA patients which may have implication in the disease pathobiology.


Anemia, Aplastic , Bone Marrow , Humans , Bone Marrow/metabolism , Bone Marrow/pathology , Anemia, Aplastic/metabolism , Anemia, Aplastic/pathology , Metabolomics , Magnetic Resonance Spectroscopy , Biomarkers , Acetates , Hypoxanthines
4.
Front Immunol ; 14: 1213560, 2023.
Article En | MEDLINE | ID: mdl-37818364

Poor graft function (PGF), manifested by multilineage cytopenias and complete donor chimerism post-allogeneic stem cell transplantation (alloSCT), and acquired aplastic anaemia (AA) are immune-mediated acquired bone marrow (BM) failure syndromes with a similar clinical presentation. In this study, we used spatial proteomics to compare the immunobiology of the BM microenvironment and identify common mechanisms of immune dysregulation under these conditions. Archival BM trephines from patients exhibited downregulation of the immunoregulatory protein VISTA and the M2 macrophage marker and suppressor of T-cell activation ARG1 with increased expression of the immune checkpoint B7-H3 compared to normal controls. Increased CD163 and CD14 expression suggested monocyte/macrophage skewing, which, combined with dysregulation of STING and VISTA, is indicative of an environment of reduced immunoregulation resulting in the profound suppression of hematopoiesis in these two conditions. There were no changes in the immune microenvironment between paired diagnostic AA and secondary MDS/AML samples suggesting that leukaemic clones develop in the impaired immune microenvironment of AA without the need for further alterations. Of the eight proteins with dysregulated expression shared by diagnostic AA and PGF, the diagnostic AA samples had a greater fold change in expression than PGF, suggesting that these diseases represent a spectrum of immune dysregulation. Unexpectedly, analysis of samples from patients with good graft function post-alloSCT demonstrated significant changes in the immune microenvironment compared to normal controls, with downregulation of CD44, STING, VISTA, and ARG1, suggesting that recovery of multilineage haematopoiesis post-alloSCT does not reflect recovery of immune function and may prime patients for the development of PGF upon further inflammatory insult. The demonstrable similarities in the immunopathology of AA and PGF will allow the design of clinical interventions that include both patient cohorts to accelerate therapeutic discovery and translation.


Anemia, Aplastic , Hematopoietic Stem Cell Transplantation , Pancytopenia , Humans , Proteomics , Bone Marrow , Bone Marrow Failure Disorders , Anemia, Aplastic/metabolism
5.
Stem Cell Res Ther ; 14(1): 285, 2023 10 04.
Article En | MEDLINE | ID: mdl-37794484

BACKGROUND: Previous studies have verified the dysfunction of mesenchymal stem cells (MSCs) for immunoregulation in acquired aplastic anemia (AA) patients. Exosomes derived from MSCs can partially substitute MSCs acting as immune regulator. Dysfunction of exosomes (Exos) derived from AA-MSC (AA-Exos) may play a key role in immunologic dissonance. METHOD: In this study, CD3 + T cells were collected and cocultured with AA-Exos and exosomes derived from HD-MSC (HD-Exos). The proliferation, differentiation and activation of CD3 + T cells were detected to compare the immunosuppressive effects between AA-Exos and HD-Exos. An immune-mediated murine model of AA was structured to compare the therapeutic effect of AA-Exos and HD-Exos. Furthermore, total RNA including miRNA from exosomes we purified and total RNA of CD3 + T cells were extracted for RNA-seq in order to construct the miRNA-mRNA network for interactions and functional analysis. RESULTS: AA-Exos had impaired inhibition effects on CD3 + T cells in terms of cell proliferation, activation and differentiation compared with exosomes from HD-Exos. HD-Exos showed a more effective rescue of AA mice compared to AA-Exos. Importantly, we found some differentially expressed miRNA involved in immune response, such as miR-199, miR-128 and miR-486. The Gene Ontology analysis of differentially expressed genes (DEGs) revealed involvement of various cellular processes, such as lymphocyte chemotaxis, lymphocyte migration and response to interferon-gamma. The Kyoto Encyclopedia of Genes and Genomes analysis illustrated upregulation of critical pathways associated with T cell function after coculturing with AA-Exos compared with HD-Exos, such as graft-versus-host disease, Th17 cell differentiation and JAK-STAT signaling pathway. A miRNA-mRNA network was established to visualize the interaction between them. CONCLUSION: In summary, AA-Exos had impaired immunosuppressive effect on T cells, less ability to rescue AA mice and differently expressed miRNA profile, which might partly account for the pathogenesis of AA as well as provide a new target of AA treatment.


Anemia, Aplastic , Exosomes , Mesenchymal Stem Cells , MicroRNAs , Humans , Mice , Animals , Exosomes/metabolism , Anemia, Aplastic/genetics , Anemia, Aplastic/therapy , Anemia, Aplastic/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mesenchymal Stem Cells/metabolism , RNA, Messenger/metabolism
6.
Sci Transl Med ; 15(678): eabo0205, 2023 01 11.
Article En | MEDLINE | ID: mdl-36630481

The common γ chain (γc; IL-2RG) is a subunit of the interleukin (IL) receptors for the γc cytokines IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. The lack of appropriate neutralizing antibodies recognizing IL-2RG has made it difficult to thoroughly interrogate the role of γc cytokines in inflammatory and autoimmune disease settings. Here, we generated a γc cytokine receptor antibody, REGN7257, to determine whether γc cytokines might be targeted for T cell-mediated disease prevention and treatment. Biochemical, structural, and in vitro analysis showed that REGN7257 binds with high affinity to IL-2RG and potently blocks signaling of all γc cytokines. In nonhuman primates, REGN7257 efficiently suppressed T cells without affecting granulocytes, platelets, or red blood cells. Using REGN7257, we showed that γc cytokines drive T cell-mediated disease in mouse models of graft-versus-host disease (GVHD) and multiple sclerosis by affecting multiple aspects of the pathogenic response. We found that our xenogeneic GVHD mouse model recapitulates hallmarks of acute and chronic GVHD, with T cell expansion/infiltration into tissues and liver fibrosis, as well as hallmarks of immune aplastic anemia, with bone marrow aplasia and peripheral cytopenia. Our findings indicate that γc cytokines contribute to GVHD and aplastic anemia pathology by promoting these characteristic features. By demonstrating that broad inhibition of γc cytokine signaling with REGN7257 protects from immune-mediated disorders, our data provide evidence of γc cytokines as key drivers of pathogenic T cell responses, offering a potential strategy for the management of T cell-mediated diseases.


Anemia, Aplastic , Graft vs Host Disease , Interleukin Receptor Common gamma Subunit , T-Lymphocytes , Animals , Mice , Anemia, Aplastic/metabolism , Antibodies, Monoclonal/metabolism , Cytokines/metabolism , Graft vs Host Disease/metabolism , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Interleukin Receptor Common gamma Subunit/antagonists & inhibitors , Interleukin Receptor Common gamma Subunit/metabolism , Primates
7.
Clin Nucl Med ; 47(12): 1048-1055, 2022 Dec 01.
Article En | MEDLINE | ID: mdl-36190714

PURPOSE: Peripheral cytopenias are typical of blood test abnormalities associated with a variety of conditions, including aplastic anemia (AA) and myelodysplastic syndromes (MDSs). We prospectively investigated the feasibility of quantitative analysis of whole-body bone marrow activity using PET with 3'-deoxy-3'- 18 F-fluorothymidine ( 18 F-FLT) in AA and MDS. PATIENTS AND METHODS: Sixty-eight patients with cytopenia underwent 18 F-FLT PET/MRI scan, with simultaneous bone marrow aspiration and biopsy for hematopoiesis evaluation. SUVs were measured in the vertebrae (Th3, 6, and 9 and L3), bilateral iliac crests, and extremities. SUV and bone marrow pathology were compared between AA and MDS and analyzed in relation to severity of AA and prognosis of MDS. RESULTS: Of the 68 patients with cytopenia, 12 were diagnosed with AA, 27 with MDS, 12 with bone marrow neoplasia, 2 with myelofibrosis, and 15 with other conditions. Iliac 18 F-FLT SUVs were significantly correlated with bone marrow cell numbers and cell density ( r = 0.47, P < 0.001 and ρ = 0.65, P < 0.001, respectively). There was a significant positive correlation between iliac and vertebral SUVs in AA and MDS ( r = 0.65, P < 0.05 and r = 0.70, P < 0.001, respectively), and the slope of the regression line was significantly steeper in AA than in MDS ( P < 0.05). In AA patients, vertebral 18 F-FLT SUVs significantly decreased with disease progression, and in MDS patients, higher whole-body 18 F-FLT uptake was associated with shorter overall survival (hazards ratio, 3.18; 95% confidence interval, 1.07-9.47; P = 0.037). CONCLUSIONS: Quantitative whole-body bone marrow imaging using 18 F-FLT PET helps distinguish AA from MDS and assess the severity of AA and prognosis of MDS.


Anemia, Aplastic , Myelodysplastic Syndromes , Humans , Anemia, Aplastic/diagnostic imaging , Anemia, Aplastic/metabolism , Bone Marrow/pathology , Myelodysplastic Syndromes/diagnostic imaging , Positron-Emission Tomography/methods , Bone Marrow Cells/metabolism
8.
Cell Biol Int ; 46(11): 1970-1976, 2022 Nov.
Article En | MEDLINE | ID: mdl-35998254

Mesenchymal stromal cells (MSC) regulate hematopoiesis in the bone marrow (BM) niche and extracellular vesicles (EVs) released by BM-MSC are important mediators of the cross-talk between BM-MSC and hematopoietic stem and progenitor cells (HSPC). We have previously demonstrated that BM-MSC of severe aplastic anemia (SAA) patients have an altered expression of hematopoiesis regulatory molecules. In the present study, we observed that CD34+ HSPC when cocultured with BM-MSC EVs from aplastic anemia patients exhibited a significant reduction in colony-forming units (p = .001), cell proliferation (p = .002), and increased apoptosis (p > .001) when compared to coculture with BM-MSC EVs from controls. Collectively, our results highlight that EVs derived from the BM-MSC of SAA patients impair the hematopoiesis supporting function of HSPC.


Anemia, Aplastic , Extracellular Vesicles , Mesenchymal Stem Cells , Anemia, Aplastic/metabolism , Antigens, CD34/metabolism , Bone Marrow , Bone Marrow Cells , Hematopoietic Stem Cells , Humans
9.
Haematologica ; 107(12): 2834-2845, 2022 12 01.
Article En | MEDLINE | ID: mdl-35734923

Myeloid-derived suppressor cells (MDSC) are a group of heterogeneous immature myeloid cells and display immunosuppressive function. In this study, MDSC populations were evaluated in acquired aplastic anemia (AA) (n=65) in which aberrant immune mechanisms contributed to bone marrow destruction. Our data demonstrate that both the proportion and immunosuppressive function of MDSC are impaired in AA patients. Decreased percentage of MDSC, especially monocytic MDSC, in the blood of AA patients (n=15) is positively correlated with the frequency of T-regulatory cells, bone marrow level of WT1 and decreased plasma level of arginase-1. RNA sequencing analyses reveal that multiple pathways including DNA damage, interleukin 4, apoptosis, and Jak kinase singnal transducer and activator of transcription are upregulated, whereas transcription, IL-6, IL-18, glycolysis, transforming growth factor and reactive oxygen species are downregulated in MDSC of AA (n=4), compared with that of healthy donors (n=3). These data suggest that AA MDSC are defective. Administration of rapamycin significantly increases the absolute number of MDSC and levels of intracellular enzymes, including arginase-1 and inducible nitric-oxide synthase. Moreover, rapamycin inhibits MDSC from differentiating into mature myeloid cells. These findings reveal that impaired MDSC are involved in the immunopathogenesis of AA. Pharmacologically targeting of MDSC by rapamycin might provide a promising therapeutic strategy for AA.


Anemia, Aplastic , Myeloid-Derived Suppressor Cells , Humans , Myeloid-Derived Suppressor Cells/metabolism , Arginase/genetics , Anemia, Aplastic/metabolism , Cell Differentiation , Immunosuppressive Agents , Sirolimus/pharmacology
10.
Mol Biol Rep ; 49(9): 8317-8324, 2022 Sep.
Article En | MEDLINE | ID: mdl-35708859

BACKGROUND: Icariin, the main pharmacological active flavonoid extracted from Epimedi herba, can regulate cellular processes in diverse diseases. The aim of this study was to explore the effects and mechanisms of icariin on proliferation and adipogenesis of bone marrow mesenchymal stem cells (BMSCs) in aplastic anemia (AA). METHODS AND RESULTS: Bone marrow mesenchymal stem cells were isolated from posterior tibias and femurs of AA rats that were induced by benzene and cyclophosphamide and gavaged with icariin. The isolated BMSCs were characterized morphologically and immunologically by positive markers (CD29 and CD90) and negative markers (CD34 and CD45). CCK-8 assay was performed to examine the BMSCs proliferation. Cell apoptosis and cell cycle were detected by flow cytometry. Oil red O staining was carried out to evaluate the adipogenesis of BMSCs. The mRNA expression of PPARγ, C/EBP-α, and FABP4 was measured by qRT-PCR. The protein levels of p-p38/p38, p-JNK/JNK, p-ERK/ERK, PPARγ, C/EBP-α, and FABP4 were detected using Western blotting. Icariin promoted the proliferation of BMSCs from AA rats in a dose-dependent manner. The protein levels of p-p38/p38, p-JNK/JNK, and p-ERK/ERK were downregulated in BMSCs from AA rats after icariin treatment. Icariin inhibited the apoptosis and arrested cell cycle at G/S phase of BMSCs from AA rats. The adipogenesis of BMSCs from AA rats was also suppressed after icariin treatment. However, the effects of icariin on BMSCs were weakened by p38 agonist addition. CONCLUSIONS: Icariin promoted the proliferation and inhibited the apoptosis and adipogenesis of BMSCs in AA by suppressing MAPK pathway.


Anemia, Aplastic , Mesenchymal Stem Cells , Anemia, Aplastic/drug therapy , Anemia, Aplastic/metabolism , Animals , Bone Marrow Cells , Cell Differentiation , Cells, Cultured , Flavonoids/metabolism , Flavonoids/pharmacology , Mesenchymal Stem Cells/metabolism , PPAR gamma/metabolism , Rats
11.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 30(3): 824-831, 2022 Jun.
Article Zh | MEDLINE | ID: mdl-35680812

OBJECTIVE: To explore the improvement effect of CXC chemokine receptor 4 (Cxcr4) gene-modified bone marrow mesenchymal stem cell (BMSC)-derived exosomes on aplastic anemia (AA), and make a preliminary exploration of the mechanism. METHODS: Mouse BMSCs were isolated and cultured, then infected by recombinant lentivirus carrying Cxcr4 gene. The expression of green fluorescence was observed through fluorescence microscope, the expression of Cxcr4 mRNA was detected by real-time fluorescence quantitative PCR, and the BMSC-derived exosomes modified with Cxcr4 gene were extracted. Mouse models of AA were constructed, and control group, model group (AA), AA+BMSC group, AA+NC-BMSC group, AA+Cxcr4-BMSC group were set up. Except control group and model group, the other three groups of mice were injected 400 µl exosomes from different sources via the tail vein, after 2 weeks, the routine blood indices and the number of bone marrow nucleated cells were detected, the pathological changes of bone marrow were observed by HE staining, and the expression level of Treg cells was detected by flow cytometry. RESULTS: Mouse BMSCs were successfully isolated, and BMSCs with high expression of Cxcr4 and their exosomes were obtained. Compared with the control group, the number of red blood cell (RBC), white blood cell (WBC), and platelet (PLT), the hemoglobin (Hb) content and proportion of Treg cells in the peripheral blood of mice in the model group significantly decreased (P<0.01), as well as the number of bone marrow nucleated cells (P<0.01). The proliferation level of nucleated cells was low, and the medullary cavity was filled with a large number of fat cells. Compared with the model group, the number of RBC, WBC, PLT, the Hb content and proportion of Treg cells in the peripheral blood of mice in the AA+BMSC group, AA+NC-BMSC group, and AA+Cxcr4-BMSC group significantly increased (P<0.01), as well as the number of bone marrow nucleated cells (P<0.01), and pathological changes of bone marrow were improved. In addition, the number of RBC, WBC, PLT, the Hb content and proportion of Treg cells in the peripheral blood of mice in the AA+Cxcr4-BMSC group were significantly higher than those in the AA+BMSC group (P<0.01), as well as the number of bone marrow nucleated cells (P<0.01). CONCLUSION: Injection of Cxcr4 gene-modified BMSC-derived exosomes has a certain improvement effect on AA mice, and the mechanism may be related to an increase of the proportion of Treg cells.


Anemia, Aplastic , Exosomes , Mesenchymal Stem Cells , Anemia, Aplastic/metabolism , Animals , Bone Marrow Cells , Exosomes/metabolism , Humans , Mice , Receptors, CXCR4
12.
Pharm Biol ; 60(1): 1160-1168, 2022 Dec.
Article En | MEDLINE | ID: mdl-35695011

CONTEXT: The effects of Rhodiola rosea L. (Crassulaceae) polysaccharides (RRPs) on haematopoiesis are poorly understood. OBJECTIVE: To determine the effects of RRPs on haematopoiesis in mice with aplastic anaemia. MATERIALS AND METHODS: Aplastic anaemia was induced in Kunming mice by 60Coγ (2.0 Gy) irradiation and cyclophosphamide administration (50 mg/kg/day for 3 consecutive days; intraperitoneal injection). The in vivo effects of RRPs (10, 20, and 40 mg/kg; intraperitoneal injection) on haematopoiesis were analyzed using peripheral blood tests, histopathological examination of haematopoietic tissues, culture of haematopoietic progenitors and bone marrow stromal cells (BMSCs), and Western blotting of Fas and Fas ligand (FasL). The in vitro effects of RRPs on bone-marrow haematopoietic progenitors and BMSCs were also evaluated. RESULTS: Compared to anaemic controls, high-dose RRPs (40 mg/kg) significantly increased red blood cells (8.21 ± 0.57835 versus 6.13 ± 1.34623 × 1012/L), white blood cells (5.11 ± 1.6141 versus l.54 ± 1.1539 × 109/L), and BMSCs (10.33 ± 1.5542 versus 5.87 ± 3.1567 × 1012/L) in mice with aplastic anaemia (all p < 0.01). High-dose RRPs significantly increased the formation of colony-forming unit-granulocyte macrophage (CFU-GM), burst-forming unit-erythroid (BFU-E), and colony-forming unit-erythroid (CFU-E; p < 0.01). Fas and FasL protein expression in BMSCs decreased after RRPs administration. Especially at the high dose, RRPs (150 µg/mL) significantly promoted in vitro CFUs-E, BFUs-E, and CFUs-GM formation. RRPs (150-300 µg/mL) also promoted BMSC proliferation. DISCUSSION AND CONCLUSIONS: RRPs helped to promote haematopoietic recovery in mice with aplastic anaemia, facilitating haematopoietic tissue recovery. This study indicated some mechanisms of the haematopoietic regulatory effects of RRPs. Our findings provide a laboratory basis for clinical research on RRPs.


Anemia, Aplastic , Rhodiola , Anemia, Aplastic/drug therapy , Anemia, Aplastic/metabolism , Animals , Bone Marrow , Bone Marrow Cells , Cell Proliferation , Colony-Forming Units Assay , Hematopoietic Stem Cells , Mice , Polysaccharides/metabolism , Polysaccharides/pharmacology , Stromal Cells
13.
Dis Markers ; 2022: 3700691, 2022.
Article En | MEDLINE | ID: mdl-35571618

Severe aplastic anemia (SAA) is an immune-mediated bone marrow failure characterized by pancytopenia. This study was aimed at uncovering proteins of plasma that were differentially expressed in SAA patients. 8 SAA patients and 8 health controls were enrolled and detected by data independent acquisition (DIA) technology. 154 differential expression proteins (DEPs) in plasma of SAA patients were identified. GO and KEGG analyses indicated DEPs were mainly involved in the immune system process. Specifically, C-C motif chemokine 18 (CCL18), matrix metalloproteinase-3 (MMP3), histidine-rich glycoprotein (HRG), and lactotransferrin (lactoferrin (Lf)) may play an important role in the immune pathogenesis of SAA. CCL18, MMP3, HRG, and Lf might be potential biomarkers for SAA.


Anemia, Aplastic , Anemia, Aplastic/genetics , Anemia, Aplastic/metabolism , Humans , Matrix Metalloproteinase 3 , Proteomics
14.
J Cell Mol Med ; 26(12): 3460-3470, 2022 06.
Article En | MEDLINE | ID: mdl-35579089

Cofilin-1 interacts with actin to regulate cell movement. The importance of cofilin-1 in immunity has been established, and its involvement in a number of autoimmune diseases has been confirmed. However, its role in severe aplastic anaemia (SAA) remains elusive. Thus, the aim of the current study was to investigate the role of cofilin-1 in patients with SAA. Flow cytometry, Western blotting and real-time quantitative reverse transcription-polymerase chain reaction were performed to detect the mRNA and protein expression of cofilin-1 in myeloid dendritic cells (mDCs) from patients with SAA. The expression of cofilin-1 was then suppressed via siRNA, and its effects on mDCs and downstream effector T-cell function were evaluated. Cofilin-1 expression was higher in mDCs from patients with SAA and correlated with routine blood and immune indexes. Moreover, cofilin-1 knockdown in mDCs from patients with SAA reduced their phagocytic capacity, migration capacity, and CD86 expression through F-actin remodelling, downregulating the stimulatory capacity of mDCs on CD4+ and CD8+ T lymphocytes. Collectively, these findings indicate that cofilin-1 participates in the hyperfunction of mDCs in patients with SAA and that the downregulation of cofilin-1 in mDCs from patients with SAA could be a novel treatment approach for SAA.


Anemia, Aplastic , Anemia, Aplastic/genetics , Anemia, Aplastic/metabolism , Dendritic Cells , Flow Cytometry , Humans , Lymphocyte Count , T-Lymphocytes/metabolism
15.
Front Immunol ; 13: 810620, 2022.
Article En | MEDLINE | ID: mdl-35450072

Thrombocytopenia is a multifactorial condition that frequently involves concomitant defects in platelet production and clearance. The physiopathology of low platelet count in thrombocytopenia remains unclear. Sialylation on platelet membrane glycoprotein and follicular helper T cells (TFHs) are thought to be the novel platelet clearance pathways. The aim of this study was to clarify the roles of platelet desialylation and circulating TFHs in patients with immune thrombocytopenia (ITP) and non-ITP thrombocytopenia. We enrolled 190 patients with ITP and 94 patients with non-ITP related thrombocytopenia including case of aplastic anemia (AA) and myelodysplastic syndromes (MDS). One hundred and ten healthy volunteers were included as controls. We found significantly increased desialylated platelets in patients with ITP or thrombocytopenia in the context of AA and MDS. Platelet desialylation was negatively correlated with platelet count. Meanwhile, the circulating TFH levels in patients with thrombocytopenia were significantly higher than those of normal controls, and were positively correlated with desialylated platelet levels. Moreover, TFHs-related chemokine CXCL13 and apoptotic platelet levels were abnormally high in ITP patients. The upregulation of pro-apoptotic proteins and the activation of the MAPK/mTOR pathway were observed in the same cohort. These findings suggested that platelet desialylation and circulating TFHs may become the potential biomarkers for evaluating the disease process associated with thrombocytopenia in patients with ITP and non-ITP.


Anemia, Aplastic , Myelodysplastic Syndromes , Purpura, Thrombocytopenic, Idiopathic , Thrombocytopenia , Anemia, Aplastic/metabolism , Blood Platelets , Humans , Myelodysplastic Syndromes/metabolism , Platelet Count , T Follicular Helper Cells , Thrombocytopenia/metabolism
16.
Front Immunol ; 13: 859668, 2022.
Article En | MEDLINE | ID: mdl-35355996

Background: In most patients with aplastic anemia (AA), the diagnosis is limited to a description of the symptoms. Lack of understanding of the underlying pathophysiological mechanisms causing bone marrow failure (BMF), hampers tailored treatment. In these patients, auto-immune cell-mediated destruction of the bone marrow is often presumed to be the causative mechanism. The status of the bone marrow microenvironment, particularly the mesenchymal stromal cell (MSC) component, was recently suggested as a potential player in the pathophysiology of AA. Therefore, functional, and immune modulatory characteristics of bone marrow MSCs might represent important parameters for AA. Objective: To conduct a systematic review to evaluate in vitro functional properties of MSCs derived from patients with AA compared to healthy controls. Methods: According to PRISMA guidelines, a comprehensive search strategy was performed by using online databases (Pubmed, ISI Web of Science, Embase, and the Cochrane Library). Studies reporting on phenotypical characterization, proliferation potential, differentiation capacity, immunomodulatory potential, and ability to support hematopoiesis were identified and screened using the Rayyan software tool. Results: 23 articles were included in this systematic review, describing a total of 324 patients with AA and 285 controls. None of the studies identified a significant difference in expression of any MSC surface marker between both groups. However, AA-MSCs showed a decreased proliferation potential, an increased tendency to differentiate into the adipogenic lineage and decreased propensity towards osteogenic differentiation. Importantly, AA-MSCs show reduced capacity of immunosuppression and hematopoietic support in comparison to healthy controls. Conclusion: We conclude that there are indications for a contribution of MSCs in the pathophysiology of AA. However, the current evidence is of poor quality and requires better defined study populations in addition to a more robust methodology to study MSC biology at a cellular and molecular level. Future studies on bone marrow microenvironment should aim at elucidating the interaction between MSCs, hematopoietic stem cells (HSCs) and immune cells to identify impairments associated with/causing BMF in patients with AA.


Anemia, Aplastic , Mesenchymal Stem Cells , Anemia, Aplastic/metabolism , Bone Marrow/metabolism , Cell Differentiation , Humans , Mesenchymal Stem Cells/metabolism , Osteogenesis
17.
Cytokine ; 152: 155829, 2022 04.
Article En | MEDLINE | ID: mdl-35217430

Acquired aplastic anemia (AA), a paradigm of bone marrow failure syndrome, is mainly caused by abnormal immune activation. The enhanced adipogenesis of bone marrow-derived mesenchymal stem cell (BM-MSC) results in a fatty marrow of AA. Leptin, an adipokine mainly generated by adipocytes, has powerful proinflammatory effects on immune cells and is associated with various autoimmune diseases. However, the role of leptin in the hyperimmune status of AA remains unknown. In this study, we firstly discovered the higher leptin concentration in AA-BM than that in healthy donors (HD)-BM and myelodysplastic syndrome (MDS)-BM. Then, we found AA-MSC could express high amounts of leptin during the process of adipogenesis. Compared with HD, the leptin receptor was also highly expressed on T cells in AA-BM. Furthermore, leptin significantly accelerated the proliferation and activation of T cells in AA-BM. And, leptin promoted the production of interferon-γby T cells in AA-BM. However, leptin remarkably inhibited the conversion of CD4+CD25- T cells into CD4+Foxp3+ T cells. Finally, we detected the cell signaling pathway in T cells from AA patients and found leptin could activate the STAT3 pathway. In summary, our data revealed the high expression of adipokine leptin in AA-BM which shaped a proinflammatory environment for T cells in AA-BM by activating the JAK2/STAT3 pathway.


Anemia, Aplastic , Mesenchymal Stem Cells , Anemia, Aplastic/metabolism , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Humans , Leptin/metabolism , Mesenchymal Stem Cells/metabolism
18.
J Clin Invest ; 132(4)2022 02 15.
Article En | MEDLINE | ID: mdl-35085104

Eltrombopag, an FDA-approved non-peptidyl thrombopoietin receptor agonist, is clinically used for the treatment of aplastic anemia, a disease characterized by hematopoietic stem cell failure and pancytopenia, to improve platelet counts and stem cell function. Eltrombopag treatment results in a durable trilineage hematopoietic expansion in patients. Some of the eltrombopag hematopoietic activity has been attributed to its off-target effects, including iron chelation properties. However, the mechanism of action for its full spectrum of clinical effects is still poorly understood. Here, we report that eltrombopag bound to the TET2 catalytic domain and inhibited its dioxygenase activity, which was independent of its role as an iron chelator. The DNA demethylating enzyme TET2, essential for hematopoietic stem cell differentiation and lineage commitment, is frequently mutated in myeloid malignancies. Eltrombopag treatment expanded TET2-proficient normal hematopoietic stem and progenitor cells, in part because of its ability to mimic loss of TET2 with simultaneous thrombopoietin receptor activation. On the contrary, TET inhibition in TET2 mutant malignant myeloid cells prevented neoplastic clonal evolution in vitro and in vivo. This mechanism of action may offer a restorative therapeutic index and provide a scientific rationale to treat selected patients with TET2 mutant-associated or TET deficiency-associated myeloid malignancies.


Anemia, Aplastic , Benzoates/pharmacology , Cell Proliferation , DNA-Binding Proteins , Dioxygenases , Hematopoietic Stem Cells/enzymology , Hydrazines/pharmacology , Pyrazoles/pharmacology , Anemia, Aplastic/drug therapy , Anemia, Aplastic/genetics , Anemia, Aplastic/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases/antagonists & inhibitors , Dioxygenases/genetics , Dioxygenases/metabolism , Humans , Mice , Mice, Knockout
19.
Leukemia ; 36(3): 834-846, 2022 03.
Article En | MEDLINE | ID: mdl-34635784

FZR1 has been implicated as a master regulator of the cell cycle and quiescence, but its roles and molecular mechanisms in the pathogenesis of severe aplastic anemia (SAA) are unclear. Here, we report that FZR1 is downregulated in SAA HSCs compared with healthy control and is associated with decreased quiescence of HSC. Haploinsufficiency of Fzr1 shows impaired quiescence and self-renewal ability of HSC in two Fzr1 heterozygous knockout mouse models. Mechanistically, FZR1 insufficiency inhibits the ubiquitination of RUNX1 protein at lysine 125, leading to the accumulation of RUNX1 protein, which disturbs the quiescence of HSCs in SAA patients. Moreover, downregulation of Runx1 reversed the loss of quiescence and impaired long-term self-renew ability in Fzr1+/- HSCs in vivo and impaired repopulation capacity in BM from SAA patients in vitro. Our findings, therefore, raise the possibility of a decisive role of the FZR1-RUNX1 pathway in the pathogenesis of SAA via deregulation of HSC quiescence.


Anemia, Aplastic/metabolism , Cdh1 Proteins/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Adult , Aged , Anemia, Aplastic/genetics , Animals , Cdh1 Proteins/genetics , Cells, Cultured , Cellular Senescence , Core Binding Factor Alpha 2 Subunit/genetics , Down-Regulation , Female , Haploinsufficiency , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Proteolysis , Ubiquitin/metabolism , Ubiquitination , Young Adult
20.
Mol Med Rep ; 25(1)2022 01.
Article En | MEDLINE | ID: mdl-34821370

Aplastic anemia (AA) is a bone marrow failure syndrome with high morbidity and mortality. Bone marrow (BM)­mesenchymal stem cells (MSCs) are the main components of the BM microenvironment, and dysregulation of BM­MSC adipogenic differentiation is a pathologic hallmark of AA. MicroRNAs (miRNAs/miRs) are crucial regulators of multiple pathological processes such as AA. However, the role of miR­30a­5p in the modulation of BM­MSC adipogenic differentiation in AA remains unclear. The present study aimed to explore the effect of miR­30a­5p on AA BM­MSC adipogenic differentiation and the underlying mechanism. The levels of miR­30a­5p expression and family with sequence similarity 13, member A (FAM13A) mRNA expression in BM­MSCs were quantified using reverse transcription­quantitative (RT­q) PCR. The mRNA expression levels of adipogenesis­associated factors [fatty acid­binding protein 4 (FABP4), lipoprotein lipase (LPL), perilipin­1 (PLIN1), peroxisome proliferator­activated receptor γ (PPARγ) and CCAAT/enhancer binding protein α (C/EBPα)] were analyzed using RT­qPCR. Lipid droplet accumulation was evaluated using Oil Red O staining in BM­MSCs. The interaction between miR­30a­5p and the FAM13A 3'­untranslated region was identified by TargetScan, and a dual­luciferase reporter assay was used to confirm the interaction. The expression levels of FAM13A and Wnt/ß­catenin pathway­related proteins were examined via western blotting. The results showed that miR­30a­5p expression levels were significantly elevated in BM­MSCs from patients with AA compared with those in control subjects (iron deficiency anemia). miR­30a­5p expression levels were also significantly increased in adipose­induced BM­MSCs in a time­dependent manner. miR­30a­5p significantly promoted AA BM­MSC adipogenic differentiation, and significantly enhanced the mRNA expression levels of FABP4, LPL, PLIN1, PPARγ and C/EBPα as well as lipid droplet accumulation. miR­30a­5p was also demonstrated to target FAM13A in AA BM­MSCs. FAM13A significantly reduced BM­MSC adipogenic differentiation by activating the Wnt/ß­catenin signaling pathway. In conclusion, miR­30a­5p was demonstrated to serve a role in AA BM­MSC adipogenic differentiation by targeting the FAM13A/Wnt/ß­catenin signaling pathway. These findings suggest that miR­30a­5p may be a therapeutic target for AA.


Adipogenesis/drug effects , Anemia, Aplastic/metabolism , Cell Differentiation/drug effects , GTPase-Activating Proteins/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/pharmacology , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , Adipose Tissue , Adult , Anemia, Aplastic/genetics , Bone Marrow Cells , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Fatty Acid-Binding Proteins/metabolism , Female , GTPase-Activating Proteins/genetics , Humans , Iron Deficiencies , Male , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , PPAR gamma/metabolism , Wnt Signaling Pathway/genetics , Young Adult
...