Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.459
Filter
1.
Eur J Pharmacol ; 978: 176793, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38960061

ABSTRACT

In recent years, a common-used antidiabetic drug, liraglutide, was identified with extra effects on lipid metabolism. Its effects against excessive lipid deposition in bone marrow were gained much attention but not well established. Our aim in the present study is to explore the interaction of miRNAs-mRNAs altered by liraglutide administration during bone marrow adipogenesis in diabetes. To establish the diabetic animal model, rats were treated with high fat diet (HFD) and STZ injection. We then identified the lowering effect of liraglutide on lipids metabolism in the diabetes. During this process, high-throughput sequencing and bioinformatics analyses on miRNAs extracted from bone marrow mesenchymal stem cells (BMSCs) were conducted after liraglutide administration. We then identified five differentially expressed miRNAs (miRNA-150-5p, miRNA-129-5p, miRNA-201-3p, miRNA-201-5p, and miRNA-214-5p). The expressions of the DE miRNAs were verified as temporal specific expression patterns in Day 3 and in Day 7. Among them, miRNA-150-5p expression was more stable and consistent with the sequencing data. Of interest, miR-150-5p overexpression facilitated adipogenesis of BMSCs. But this promotion was alleviated by liraglutide. The predicted target gene of miR-150-5p, GDF11, was validated to be involved in liraglutide alleviated BMSCs' lipid accumulation in diabetes. In vitro, liraglutide increased the GDF11 expression, rescued its down-expression by siGDF11 and inhibit the adipogenesis of BMSCs cultured in high glucose medium. In vivo, liraglutide reversed the HFD-STZ induced excessive lipid droplets by up-regulation of GDF11 expression, which was discounted by agomiR-150-5p injection. Above all, liraglutide might alleviate bone marrow fat accumulation via inactivating miR-150-5p/GDF11 axis in diabetes.


Subject(s)
Adipogenesis , Diabetes Mellitus, Experimental , Liraglutide , Mesenchymal Stem Cells , MicroRNAs , Rats, Sprague-Dawley , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Liraglutide/pharmacology , Liraglutide/therapeutic use , Male , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Adipogenesis/drug effects , Adipogenesis/genetics , Rats , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Growth Differentiation Factors/genetics , Growth Differentiation Factors/metabolism , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Bone Marrow/drug effects , Bone Marrow/metabolism , Diet, High-Fat/adverse effects , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Gene Expression Regulation/drug effects , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism
2.
Bull Exp Biol Med ; 177(1): 30-34, 2024 May.
Article in English | MEDLINE | ID: mdl-38954304

ABSTRACT

Topotecan administered intraperitoneally at single doses of 0.25, 0.5, and 1 mg/kg induced chromosomal aberrations in bone marrow cells of F1(CBA×C57BL/6) hybrid mice in a dose-dependent manner. A tyrosyl-DNA phosphodiesterase 1 (TDP1) inhibitor, an usnic acid derivative OL9-116 was inactive in a dose range of 20-240 mg/kg, but enhanced the cytogenetic effect of topotecan (0.25 mg/kg) at a dose of 40 mg/kg (per os). The TDP1 inhibitor, a coumarin derivative TX-2552 (at doses of 20, 40, 80, and 160 mg/kg per os), increased the level of aberrant metaphases induced by topotecan (0.25 mg/kg) by 2.1-2.6 times, but was inactive at a dose of 10 mg/kg. The results indicate that TDP1 inhibitors enhance the clastogenic activity of topotecan in mouse bone marrow cells in vivo and are characterized by different dose profiles of the co-mutagenic effects.


Subject(s)
Bone Marrow Cells , Phosphoric Diester Hydrolases , Topotecan , Animals , Topotecan/pharmacology , Mice , Phosphoric Diester Hydrolases/metabolism , Bone Marrow Cells/drug effects , Male , Chromosome Aberrations/drug effects , Chromosome Aberrations/chemically induced , Phosphodiesterase Inhibitors/pharmacology , Topoisomerase I Inhibitors/pharmacology , Mice, Inbred C57BL , Mutagens/toxicity
3.
Mol Biol Rep ; 51(1): 748, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874843

ABSTRACT

Background this study was conducted to assess the effects of vitamin D on differentiation of bone marrow- derived mesenchymal stem cells (BM-MSCs) into insulin producing cells (IPCs). Method BM-MSCs were isolated from femur and tibia of rats and incubated in low (LG) or high glucose (HG) (5mM or 25mM), or high glucose DMEM media supplemented with vitamin D (0.2nM) (HGD) for 14 days. Cells viability was analysis by MTT assay. Differentiation of SCs was confirmed using measuring genes expression level of pdx1 and insulin, and insulin secretion, glucose stimulated insulin secretion, and insulin content by ELISA method. Results Cell viability was significantly higher in HGD than LG (p < 0.05) in day 3, also, in HG and HGD than LG (p < 0.001), and HGD vs. HG (p < 0.001) in day 7. Pdx1 and insulin level was markedly higher in HGD than LG (p < 0.05 and p < 0.01). pdx1 expression was markedly higher in HGD (p < 0.05) than LG, also insulin expression the HG (p < 0.05), and HGD (p < 0.01) groups compared to the LG group. Insulin release at 5mM glucose was notably higher in the HGD group compared to LG (p < 0.05), and at 25mM glucose, both HG and HGD showed significant increases vs. LG (p < 0.05 and p < 0.01, respectively). Insulin content was significantly higher in both 5mM and 25mM glucose for HG and HGD vs. LG (p < 0.01 and p < 0.001, respectively). In conclusion, treatment BM-MSCs with vitamin D could increase their differentiation into IPCs and it can be considered as a potential supplementary agent in enhancing differentiation SCs into insulin generating cells.


Subject(s)
Bone Marrow Cells , Cell Differentiation , Insulin-Secreting Cells , Insulin , Mesenchymal Stem Cells , Vitamin D , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Animals , Cell Differentiation/drug effects , Vitamin D/pharmacology , Vitamin D/metabolism , Rats , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/cytology , Glucose/metabolism , Glucose/pharmacology , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Cells, Cultured , Cell Survival/drug effects , Male , Trans-Activators/metabolism , Trans-Activators/genetics , Dietary Supplements , Insulin Secretion/drug effects
4.
Biosci Rep ; 44(6)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38836325

ABSTRACT

Natural and synthetic polymeric materials, particularly soft and hard tissue replacements, are paramount in medicine. We prepared calcium-incorporated sulfonated polyether-ether ketone (SPEEK) polymer membranes for bone applications. The bioactivity was higher after 21 days of immersion in simulated body fluid (SBF) due to calcium concentration in the membrane. We present a new biomaterial healing system composed of calcium and sulfonated polyether ether ketone (Ca-SPEEK) that can function as a successful biomaterial without causing inflammation when tested on bone marrow cells. The Ca-SPEEK exhibited 13 ± 0.5% clot with low fibrin mesh formation compared to 21 ± 0.5% in SPEEK. In addition, the Ca-SPEEK showed higher protein adsorption than SPEEK membranes. As an inflammatory response, IL-1 and TNF-α in the case of Ca-SPEEK were lower than those for SPEEK. We found an early regulation of IL-10 in the case of Ca-SPEEK at 6 h, which may be attributed to the down-regulation of the inflammatory markers IL-1 and TNF-α. These results evidence the innovative bioactivity of Ca-SPEEK with low inflammatory response, opening venues for bone applications.


Subject(s)
Biocompatible Materials , Bone Marrow Cells , Calcium , Polymers , Tumor Necrosis Factor-alpha , Animals , Mice , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Polymers/chemistry , Polymers/pharmacology , Calcium/metabolism , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Benzophenones/chemistry , Benzophenones/pharmacology , Inflammation/drug therapy , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Ketones/chemistry , Ketones/pharmacology , Materials Testing , Interleukin-1/metabolism , Interleukin-10/metabolism
5.
Cells ; 13(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38891064

ABSTRACT

Periodontal disease is characterized by inflammation and bone loss. Central to its pathogenesis is the dysregulated inflammatory response, complicating regenerative therapies. Mesenchymal stem cells (MSCs) hold significant promise in tissue repair and regeneration. This study investigated the effects of specialized pro-resolving mediators (SPMs), Resolvin E1 (RvE1) and Maresin 1 (MaR1), on the osteogenic differentiation of human bone marrow-derived MSCs under inflammatory conditions. The stem cells were treated with SPMs in the presence of lipopolysaccharide (LPS) to simulate an inflammatory environment. Osteogenic differentiation was assessed through alkaline phosphatase activity and alizarin red staining. Proteomic analysis was conducted to characterize the protein expression profile changes, focusing on proteins related to osteogenesis and osteoclastogenesis. Treatment with RvE1 and MaR1, both individually and in combination, significantly enhanced calcified deposit formation. Proteomic analysis revealed the differential expression of proteins associated with osteogenesis and osteoclastogenesis, highlighting the modulatory impact of SPMs on bone metabolism. RvE1 and MaR1 promote osteogenic differentiation of hBMMSCs in an inflammatory environment, with their combined application yielding synergistic effects. This study provides insights into the therapeutic potential of SPMs in enhancing bone regeneration, suggesting a promising avenue for developing regenerative therapies for periodontal disease and other conditions characterized by inflammation-induced bone loss.


Subject(s)
Cell Differentiation , Docosahexaenoic Acids , Eicosapentaenoic Acid , Inflammation , Mesenchymal Stem Cells , Osteogenesis , Osteogenesis/drug effects , Humans , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/analogs & derivatives , Docosahexaenoic Acids/pharmacology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Cell Differentiation/drug effects , Inflammation/pathology , Proteomics , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/cytology , Lipopolysaccharides/pharmacology
6.
Exp Cell Res ; 440(2): 114138, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38906316

ABSTRACT

Prolyl 4-hydroxylase beta subunit (P4HB) plays a vital role in bone formation. This study intends to clarify the role of P4HB in the therapeutic effect of Icariin (ICA) on osteoporosis. Herein, in vivo and in vitro models were constructed by performing ovariectomy (OVX) in rats and inducing osteogenic differentiation in bone marrow stem cells (BMSCs), respectively. Hematoxylin and eosin staining and micro-computed tomography analysis were performed to evaluate osteoporosis in OVX rats. Alizarin Red staining, alkaline phosphatase staining, and the ALP activity test were employed to assess osteogenesis. m6A dot blotting and methylated RNA immunoprecipitation were used to determine m6A modification. We found that P4HB was downregulated in bone tissues of patients with osteoporosis and OVX rats. P4HB facilitated osteogenic differentiation of BMSCs. What's more, ICA upregulated P4HB expression, promoted osteogenic differentiation of BMSCs, and alleviated osteoporosis in OVX rats, which were reversed by knocking down P4HB. ICA enhanced the stability and m6A modification of P4HB. METTL14 mediated m6A modification of P4HB mRNA. In addition, METTL14 knockdown overturned the promotive effects of ICA on P4HB m6A level and BMSC osteogenic differentiation. To sum up, ICA elevated the METTL14-mediated m6A modification of P4HB to facilitate BMSC osteogenic differentiation.


Subject(s)
Cell Differentiation , Flavonoids , Methyltransferases , Osteogenesis , Rats, Sprague-Dawley , Animals , Osteogenesis/drug effects , Cell Differentiation/drug effects , Rats , Female , Flavonoids/pharmacology , Methyltransferases/metabolism , Methyltransferases/genetics , Humans , Osteoporosis/pathology , Osteoporosis/metabolism , Osteoporosis/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Ovariectomy , Up-Regulation/drug effects , Procollagen-Proline Dioxygenase/metabolism , Procollagen-Proline Dioxygenase/genetics , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Cells, Cultured , Adenosine/analogs & derivatives , Adenosine/metabolism
7.
In Vivo ; 38(4): 1719-1730, 2024.
Article in English | MEDLINE | ID: mdl-38936939

ABSTRACT

BACKGROUND/AIM: ClFdA is a second-generation antineoplastic agent that has demonstrated significant anticancer activity, particularly against acute lymphoblastic leukemia and has been shown to have radiosensitizing activity. The aim of the study was to explore the genotoxic, cytotoxic and radiosensitizing effects of clofarabine (ClFdA) on bone marrow cells (BMCs), normoblasts and leukocytes of mice in vivo. MATERIALS AND METHODS: Cytotoxicity was determined by the reduction in reticulocytes (RET), and genotoxicity was determined by the induction of micronucleated reticulocytes (MN-RET) in the peripheral blood and by DNA break induction in leukocytes determined by single-cell gel electrophoresis (SCGE). The radiosensitizing capacity of ClFdA was determined in leukocytes and BMCs by SCGE. RESULTS: Two mechanisms of MN-RET induction were identified according to the antecedents, that could be due to inhibition of DNA synthesis and demethylation of G-C regions, and subsequent chromosome fragility. ClFdA cytotoxicity causes two contiguous peaks, an early peak that seems to inhibit MN-RET induction and a second peak that seems to be caused by ribonucleotide reductase (RR) and/or DNA synthesis inhibitions. ClFdA induced early DNA damage in noncycling leukocytes, and also radiosensitizes leukocytes immediately after treatment. ClFdA-ionizing radiation (IR) causes two time-dependent episodes of DNA damage, the latest after 80 min triggers a major breakage of DNA. In terms of the number of damaged cells, leukocytes and BMCs are similarly sensitive to ionizing radiation; BMCs are slightly more sensitive than leukocytes to ClFdA, but BMCs are doubly sensitive to combined treatment. CONCLUSION: ClFdA causes early DNA damage and radiosensitivity in non-proliferating leukocytes, which rules out the most favored hypotheses of the participation of RR and DNA polymerase inhibition.


Subject(s)
Clofarabine , DNA Damage , Leukocytes , Radiation-Sensitizing Agents , Animals , Clofarabine/pharmacology , Mice , Radiation-Sensitizing Agents/pharmacology , Leukocytes/drug effects , Leukocytes/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Arabinonucleosides/pharmacology , Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Bone Marrow Cells/metabolism , Adenine Nucleotides/pharmacology , Male , Reticulocytes/drug effects , Reticulocytes/radiation effects , Antineoplastic Agents/pharmacology , Micronucleus Tests
8.
Ann Hematol ; 103(8): 3015-3027, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38847852

ABSTRACT

Bone marrow stromal cells (BMSCs) can promote the growth of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). Histone deacetylases (HDACs) play essential roles in the proliferation and apoptosis resistance of Ph + ALL cells. In our previous study, inhibiting histone deacetylase 1 (HDAC1) decreases the proliferation of Ph + ALL cells. However, little is known regarding how HDAC1 in BMSCs of Ph + ALL patients affects the imatinib (IM) resistance. Therefore, the present work examined the roles of HDAC1 in BMSCs. Overexpression of HDAC1 was found in BMSCs of Ph + ALL patients with IM resistance. In addition, the Ph + ALL cell line SUP-B15 was co-cultured with BMSCs after lentivirus transfection for regulating HDAC1 expression. Knockdown of HDAC1 within BMSCs elevated the IM-mediated SUP-B15 cell apoptosis, while increasing HDAC1 expression had an opposite effect. IL-6 in BMSCs, which is an important factor for the microenvironment-associated chemoresistance, showed evident up-regulation in HDAC1-upregulated BMSCs and down-regulation in HDAC1-downregulated BMSCs. While recombinant IL-6 (rIL-6) can reversed the sensitivity of SUP-B15 cells to IM induced by downregulating HDAC1 expression in BMSCs. HDAC1 showed positive regulation on IL-6 transcription and secretion. Moreover, IL-6 secretion induced by HDAC1 in BMSCs might enhance IM resistance in Ph + ALL cells. With regard to the underlying molecular mechanism, NF-κB, an important signal responsible for IL-6 transcription in BMSCs, mediated the HDAC1-regulated IL-6 expression. Collectively, this study facilitated to develop HDAC1 inhibitors based not only the corresponding direct anti-Ph + ALL activity but also the regulation of bone marrow microenvironment.


Subject(s)
Drug Resistance, Neoplasm , Histone Deacetylase 1 , Imatinib Mesylate , Interleukin-6 , Mesenchymal Stem Cells , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Humans , Drug Resistance, Neoplasm/drug effects , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Interleukin-6/metabolism , Interleukin-6/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Male , Female , Cell Line, Tumor , Adult , Apoptosis/drug effects , Child , Adolescent , Philadelphia Chromosome , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Gene Expression Regulation, Leukemic/drug effects
9.
Mol Immunol ; 171: 93-104, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38805892

ABSTRACT

BACKGROUND: This study determines the role and mechanism of APS in cyclophosphamide-induced myelosuppression in mice and bone mesenchymal stem cells (BMSCs) cell model. METHODS: Cy-induced myelosuppression mice and BMSCs cell model were established. Fifty C57BL/6 mice (weighing 20 ± 2 g) were randomly divided into five groups. Femur and tibia samples, bone marrow samples, and blood samples were collected 3 days after the last injection of Cy. Histopathology changes and cell apoptosis were detected. Cell viability, apoptosis, cycle distribution, reactive oxygen species activity, osteogenesis ability, and protein levels were detected. γ-H2AX and senescence-associated ß-galactosidase activity expression was detected by immunofluorescence. Cy-induced senescence and Wnt/ß-catenin related protein levels were detected using western blotting. RESULTS: The results showed that APS effectively induced Cy-induced histological injury and cell apoptosis rate. After treated with APS, ROS and ALP levels were significantly increased. In BMSCs, cell viability, apoptosis, and cell cycle distribution were also influenced by APS treatment. Compared with the control group, cell viability was significantly increased, the cell apoptosis rate was decreased while the number of cells remained in the G0-G1 phase was increased. Meanwhile, ROS levels were significantly increased in APS group. Cell senescence and Wnt/ß-catenin related protein (γ-H2AX, SA-ß-gal, p21, p16, p-ß-catenin/ ß-catenin, c-Myc, and AXIN2) levels were also altered both in vivo and in vitro. Interestingly, the effects of APS were reversed by BML-284. CONCLUSION: Our results indicate that APS protected Cy-induced myelosuppression through the Wnt/ß-catenin pathway and APS is a potential therapeutic drug for Cy-induced myelosuppression.


Subject(s)
Apoptosis , Astragalus Plant , Cyclophosphamide , Mesenchymal Stem Cells , Mice, Inbred C57BL , Polysaccharides , Animals , Cyclophosphamide/toxicity , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Apoptosis/drug effects , Mice , Polysaccharides/pharmacology , Astragalus Plant/chemistry , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Wnt Signaling Pathway/drug effects , Male , beta Catenin/metabolism , Cellular Senescence/drug effects , Bone Marrow/drug effects , Bone Marrow/metabolism , Osteogenesis/drug effects , Cell Cycle/drug effects
10.
J Toxicol Environ Health A ; 87(16): 647-661, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-38804873

ABSTRACT

The present study aimed to determine the genoprotective activity and safety of Moringa oleifera leave and Tinospora cordifolia stem extracts against cyclophosphamide (CP)-induced genotoxicity utilizing Swiss albino mice. Animals were divided into 14 groups for subacute treatment with either M. oleifera or T. cordifolia extracts daily for 28 days. The extract doses selected were 100, 200 or 400 mg/kg b.w administered orally alone or combined with CP (50 mg/kg b.w. intraperitoneally daily for 5 days). Analyses performed included the comet assay, micronucleus test (MN) in bone marrow cells and sperm head abnormality assay (SHA). M. oleifera and T. cordifolia extracts induced no significant genotoxic effects on somatic and germ cells. In contrast, for all cells examined M. oleifera and T. cordifolia extracts inhibited DNA damage initiated by CP. Taken together data demonstrated that both plant extracts did not exhibit marked genotoxic effects but displayed potential chemoprotective properties against CP-induced genotoxicity in Swiss mice.


Subject(s)
Cyclophosphamide , DNA Damage , Micronucleus Tests , Moringa oleifera , Plant Extracts , Plant Leaves , Tinospora , Animals , Tinospora/chemistry , Mice , Cyclophosphamide/toxicity , Moringa oleifera/chemistry , Plant Extracts/pharmacology , Male , Plant Leaves/chemistry , DNA Damage/drug effects , Comet Assay , Plant Stems/chemistry , Bone Marrow/drug effects , Bone Marrow Cells/drug effects , Mutagens/toxicity , Antimutagenic Agents/pharmacology
11.
Cancer Lett ; 592: 216937, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38704134

ABSTRACT

Dysfunctional bone marrow (BM) endothelial progenitor cells (EPCs) with high levels of reactive oxygen species (ROS) are responsible for defective hematopoiesis in poor graft function (PGF) patients with acute leukemia or myelodysplastic neoplasms post-allotransplant. However, the underlying mechanism by which BM EPCs regulate their intracellular ROS levels and the capacity to support hematopoiesis have not been well clarified. Herein, we demonstrated decreased levels of peroxisome proliferator-activated receptor delta (PPARδ), a lipid-activated nuclear receptor, in BM EPCs of PGF patients compared with those with good graft function (GGF). In vitro assays further identified that PPARδ knockdown contributed to reduced and dysfunctional BM EPCs, characterized by the impaired ability to support hematopoiesis, which were restored by PPARδ overexpression. Moreover, GW501516, an agonist of PPARδ, repaired the damaged BM EPCs triggered by 5-fluorouracil (5FU) in vitro and in vivo. Clinically, activation of PPARδ by GW501516 benefited the damaged BM EPCs from PGF patients or acute leukemia patients in complete remission (CR) post-chemotherapy. Mechanistically, we found that increased expression of NADPH oxidases (NOXs), the main ROS-generating enzymes, may lead to elevated ROS level in BM EPCs, and insufficient PPARδ may trigger BM EPC damage via ROS/p53 pathway. Collectively, we found that defective PPARδ contributes to BM EPC dysfunction, whereas activation of PPARδ in BM EPCs improves their hematopoiesis-supporting ability after myelosuppressive therapy, which may provide a potential therapeutic target not only for patients with leukemia but also for those with other cancers.


Subject(s)
Endothelial Progenitor Cells , Hematopoiesis , PPAR delta , Reactive Oxygen Species , Adult , Animals , Female , Humans , Male , Mice , Middle Aged , Young Adult , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/drug effects , Fluorouracil/pharmacology , Hematopoiesis/drug effects , Mice, Inbred C57BL , Myelodysplastic Syndromes/pathology , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/drug therapy , NADPH Oxidases/metabolism , PPAR delta/metabolism , PPAR delta/genetics , Reactive Oxygen Species/metabolism , Thiazoles/pharmacology , Tumor Suppressor Protein p53/metabolism
12.
Stem Cell Res Ther ; 15(1): 124, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38679735

ABSTRACT

BACKGROUND: Recombinant human bone morphogenetic protein 2 (rhBMP-2) and human bone marrow mesenchymal stromal cells (hBM-MSCs) have been thoroughly studied for research and translational bone regeneration purposes. rhBMP-2 induces bone formation in vivo, and hBM-MSCs are its target, bone-forming cells. In this article, we studied how rhBMP-2 drives the multilineage differentiation of hBM-MSCs both in vivo and in vitro. METHODS: rhBMP-2 and hBM-MSCs were tested in an in vivo subcutaneous implantation model to assess their ability to form mature bone and undergo multilineage differentiation. Then, the hBM-MSCs were treated in vitro with rhBMP-2 for short-term or long-term cell-culture periods, alone or in combination with osteogenic, adipogenic or chondrogenic media, aiming to determine the role of rhBMP-2 in these differentiation processes. RESULTS: The data indicate that hBM-MSCs respond to rhBMP-2 in the short term but fail to differentiate in long-term culture conditions; these cells overexpress the rhBMP-2 target genes DKK1, HEY-1 and SOST osteogenesis inhibitors. However, in combination with other differentiation signals, rhBMP-2 acts as a potentiator of multilineage differentiation, not only of osteogenesis but also of adipogenesis and chondrogenesis, both in vitro and in vivo. CONCLUSIONS: Altogether, our data indicate that rhBMP-2 alone is unable to induce in vitro osteogenic terminal differentiation of hBM-MSCs, but synergizes with other signals to potentiate multiple differentiation phenotypes. Therefore, rhBMP-2 triggers on hBM-MSCs different specific phenotype differentiation depending on the signalling environment.


Subject(s)
Bone Morphogenetic Protein 2 , Cell Differentiation , Mesenchymal Stem Cells , Osteogenesis , Recombinant Proteins , Humans , Adipogenesis/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chondrogenesis/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
13.
Ecotoxicol Environ Saf ; 277: 116392, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38677065

ABSTRACT

Smoking disrupts bone homeostasis and serves as an independent risk factor for the development and progression of osteoporosis. Tobacco toxins inhibit the proliferation and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), promote BMSCs aging and exhaustion, but the specific mechanisms are not yet fully understood. Herein, we successfully established a smoking-related osteoporosis (SROP) model in rats and mice through intraperitoneal injection of cigarette smoke extract (CSE), which significantly reduced bone density and induced aging and inhibited osteogenic differentiation of BMSCs both in vivo and in vitro. Bioinformatics analysis and in vitro experiments confirmed that CSE disrupts mitochondrial homeostasis through oxidative stress and inhibition of mitophagy. Furthermore, we discovered that CSE induced BMSCs aging by upregulating phosphorylated AKT, which in turn inhibited the expression of FOXO3a and the Pink1/Parkin pathway, leading to the suppression of mitophagy and the accumulation of damaged mitochondria. MitoQ, a mitochondrial-targeted antioxidant and mitophagy agonist, was effective in reducing CSE-induced mitochondrial oxidative stress, promoting mitophagy, significantly downregulating the expression of aging markers in BMSCs, restoring osteogenic differentiation, and alleviating bone loss and autophagy levels in CSE-exposed mice. In summary, our results suggest that BMSCs aging caused by the inhibition of mitophagy through the AKT/FOXO3a/Pink1/Parkin axis is a key mechanism in smoking-related osteoporosis.


Subject(s)
Mesenchymal Stem Cells , Mitophagy , Osteoporosis , Animals , Mitophagy/drug effects , Mesenchymal Stem Cells/drug effects , Mice , Rats , Osteoporosis/chemically induced , Osteoporosis/pathology , Nicotiana/adverse effects , Forkhead Box Protein O3/metabolism , Oxidative Stress/drug effects , Male , Rats, Sprague-Dawley , Osteogenesis/drug effects , Cellular Senescence/drug effects , Cell Differentiation/drug effects , Smoke/adverse effects , Ubiquitin-Protein Ligases/metabolism , Mitochondria/drug effects , Protein Kinases/metabolism , Mice, Inbred C57BL , Bone Marrow Cells/drug effects
14.
Regul Toxicol Pharmacol ; 149: 105623, 2024 May.
Article in English | MEDLINE | ID: mdl-38631606

ABSTRACT

The Bone-Marrow derived Dendritic Cell (BMDC) test is a promising assay for identifying sensitizing chemicals based on the 3Rs (Replace, Reduce, Refine) principle. This study expanded the BMDC benchmarking to various in vitro, in chemico, and in silico assays targeting different key events (KE) in the skin sensitization pathway, using common substances datasets. Additionally, a Quantitative Structure-Activity Relationship (QSAR) model was developed to predict the BMDC test outcomes for sensitizing or non-sensitizing chemicals. The modeling workflow involved ISIDA (In Silico Design and Data Analysis) molecular fragment descriptors and the SVM (Support Vector Machine) machine-learning method. The BMDC model's performance was at least comparable to that of all ECVAM-validated models regardless of the KE considered. Compared with other tests targeting KE3, related to dendritic cell activation, BMDC assay was shown to have higher balanced accuracy and sensitivity concerning both the Local Lymph Node Assay (LLNA) and human labels, providing additional evidence for its reliability. The consensus QSAR model exhibits promising results, correlating well with observed sensitization potential. Integrated into a publicly available web service, the BMDC-based QSAR model may serve as a cost-effective and rapid alternative to lab experiments, providing preliminary screening for sensitization potential, compound prioritization, optimization and risk assessment.


Subject(s)
Benchmarking , Dendritic Cells , Quantitative Structure-Activity Relationship , Dendritic Cells/drug effects , Humans , Animals , Support Vector Machine , Computer Simulation , Dermatitis, Allergic Contact , Allergens/toxicity , Animal Testing Alternatives/methods , Bone Marrow Cells/drug effects , Local Lymph Node Assay , Mice
15.
FEBS Open Bio ; 14(6): 1001-1010, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38531625

ABSTRACT

Myeloperoxidase (MPO) is found almost exclusively in granulocytes and immature myeloid cells. It plays a key role in the innate immune system, catalysing the formation of reactive oxygen species that are important in anti-microbial action, but MPO also oxidatively transforms the topoisomerase II (TOP2) poison etoposide to chemical forms that have elevated DNA damaging properties. TOP2 poisons such as etoposide are widely used anti-cancer drugs, but they are linked to cases of secondary acute myeloid leukaemias through a mechanism that involves DNA damage and presumably erroneous repair leading to leukaemogenic chromosome translocations. This leads to the possibility that myeloperoxidase inhibitors could reduce the rate of therapy-related leukaemia by protecting haematopoietic cells from TOP2 poison-mediated genotoxic damage while preserving the anti-cancer efficacy of the treatment. We show here that myeloperoxidase inhibition reduces etoposide-induced TOP2B-DNA covalent complexes and resulting DNA double-strand break formation in primary ex vivo expanded CD34+ progenitor cells and unfractionated bone marrow mononuclear cells. Since MPO inhibitors are currently being developed as anti-inflammatory agents this raises the possibility that repurposing of these potential new drugs could provide a means of suppressing secondary acute myeloid leukaemias associated with therapies containing TOP2 poisons.


Subject(s)
DNA Damage , DNA Topoisomerases, Type II , Etoposide , Peroxidase , Poly-ADP-Ribose Binding Proteins , Etoposide/pharmacology , Humans , Peroxidase/metabolism , DNA Damage/drug effects , DNA Topoisomerases, Type II/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Topoisomerase II Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , DNA Breaks, Double-Stranded/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism
16.
Cell Prolif ; 57(7): e13612, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38348888

ABSTRACT

Ageing and cell senescence of mesenchymal stem cells (MSCs) limited their immunomodulation properties and therapeutic application. We previously reported that nucleosome assembly protein 1-like 2 (Nap1l2) contributes to MSCs senescence and osteogenic differentiation. Here, we sought to evaluate whether Nap1l2 impairs the immunomodulatory properties of MSCs and find a way to rescue the deficient properties. We demonstrated that metformin could rescue the impaired migration properties and T cell regulation properties of OE-Nap1l2 BMSCs. Moreover, metformin could improve the impaired therapeutic efficacy of OE-Nap1l2 BMSCs in the treatment of colitis and experimental autoimmune encephalomyelitis in mice. Mechanistically, metformin was capable of upregulating the activation of AMPK, synthesis of l-arginine and expression of inducible nitric oxide synthase in OE-Nap1l2 BMSCs, leading to an increasing level of nitric oxide. This study indicated that Nap1l2 negatively regulated the immunomodulatory properties of BMSCs and that the impaired functions could be rescued by metformin pretreatment via metabolic reprogramming. This strategy might serve as a practical therapeutic option to rescue impaired MSCs functions for further application.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Immunomodulation , Mesenchymal Stem Cells , Metformin , Mice, Inbred C57BL , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Animals , Metformin/pharmacology , Mice , Immunomodulation/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Colitis/drug therapy , Colitis/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Cellular Reprogramming/drug effects , Cell Differentiation/drug effects , Cellular Senescence/drug effects , Cells, Cultured , Cell Movement/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/cytology , Metabolic Reprogramming
17.
Int J Mol Sci ; 24(7)2023 Mar 26.
Article in English | MEDLINE | ID: mdl-37047231

ABSTRACT

The evidence supporting the biological plausibility of the association of permethrin and malathion with hematological cancer is limited and contradictory; thus, further studies are needed. This study aimed to investigate whether in vitro exposure to 0.1 µM permethrin and malathion at 0, 24, 48 and 72 h after cell culture initiation induced changes in the gene expression and DNA methylation in mononuclear cells from bone marrow and peripheral blood (BMMCs, PBMCs). Both pesticides induced several gene expression modifications in both tissues. Through gene ontology analysis, we found that permethrin deregulates ion channels in PBMCs and BMMCs and that malathion alters genes coding proteins with nucleic acid binding capacity, which was also observed in PBMCs exposed to permethrin. Additionally, we found that both insecticides deregulate genes coding proteins with chemotaxis functions, ion channels, and cytokines. Several genes deregulated in this study are potentially associated with cancer onset and development, and some of them have been reported to be deregulated in hematological cancer. We found that permethrin does not induce DNA hypermethylation but can induce hypomethylation, and that malathion generated both types of events. Our results suggest that these pesticides have the potential to modify gene expression through changes in promoter DNA methylation and potentially through other mechanisms that should be investigated.


Subject(s)
Bone Marrow Cells , DNA Methylation , Gene Expression , Insecticides , Malathion , Organophosphates , Permethrin , Gene Expression/drug effects , DNA Methylation/drug effects , Hematopoiesis/drug effects , Hematopoiesis/genetics , Permethrin/toxicity , Malathion/toxicity , Insecticides/toxicity , Organophosphates/toxicity , Bone Marrow Cells/drug effects , Blood Cells/drug effects , Humans , Male , Young Adult , Cells, Cultured
18.
Int J Mol Sci ; 24(7)2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37047701

ABSTRACT

Cartilage is an avascular tissue and sensitive to mechanical trauma and/or age-related degenerative processes leading to the development of osteoarthritis (OA). Therefore, it is important to investigate the mesenchymal cell-based chondrogenic regenerating mechanisms and possible their regulation. The aim of this study was to investigate the role of intracellular calcium (iCa2+) and its regulation through voltage-operated calcium channels (VOCC) on chondrogenic differentiation of mesenchymal stem/stromal cells derived from human bone marrow (BMMSCs) and menstrual blood (MenSCs) in comparison to OA chondrocytes. The level of iCa2+ was highest in chondrocytes, whereas iCa2+ store capacity was biggest in MenSCs and they proliferated better as compared to other cells. The level of CaV1.2 channels was also highest in OA chondrocytes than in other cells. CaV1.2 antagonist nifedipine slightly suppressed iCa2+, Cav1.2 and the proliferation of all cells and affected iCa2+ stores, particularly in BMMSCs. The expression of the CaV1.2 gene during 21 days of chondrogenic differentiation was highest in MenSCs, showing the weakest chondrogenic differentiation, which was stimulated by the nifedipine. The best chondrogenic differentiation potential showed BMMSCs (SOX9 and COL2A1 expression); however, purposeful iCa2+ and VOCC regulation by blockers can stimulate a chondrogenic response at least in MenSCs.


Subject(s)
Calcium Channel Blockers , Chondrocytes , Mesenchymal Stem Cells , Nifedipine , Osteoarthritis , Humans , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation/genetics , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrogenesis/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Nifedipine/pharmacology , Osteoarthritis/metabolism , Calcium Channels, L-Type , Calcium Channel Blockers/pharmacology
19.
Curr Mol Med ; 23(5): 410-419, 2023.
Article in English | MEDLINE | ID: mdl-35996252

ABSTRACT

BACKGROUND: Vitamin D receptor (VDR) is critical for mineral and bone homeostasis since it plays an essential role in the osteoblast differentiation of bone marrow mesenchymal stem cells (BM-MSCs). Hydroxysafflor yellow A (HSYA) has the potential to promote bone mineralization and inhibit bone resorption, while its detailed mechanism needs to be elaborated. OBJECTIVE: This study intends to explore the action of HSYA on the proliferation and differentiation of BM-MSC and the underlying mechanism. METHODS: Different concentrations of HSYA to BM-MSC and CCK-8, and EdU were used to detect cell viability and proliferation. The alkaline phosphatase (ALP) was used to observe the differentiation ability of BM-MSC osteoblasts. The calcium uptake and mineralization of osteoblast-like cells were observed by alizarin red staining. The level of calcium ion uptake in cells was detected by flow cytometry. AutoDock was performed for molecular docking of HSYA to VDR protein. Immunofluorescence and western blotting were performed to detect the expression of VDR expression levels. Finally, the effect of VDR was verified by a VDR inhibitor. RESULTS: After treatment with HSYA, the proliferation and calcium uptake of BM-MSC were increased. The level of ALP increased significantly and reached its peak on the 12th day. HSYA promoted calcium uptake and calcium deposition, and mineralization of osteoblasts. The western blotting and immunofluorescence showed that HSYA increased the expression of VDR in the osteoblast-like cell's nucleus and upregulated Osteocalcin, S100 calcium-binding protein G, and CYP24A1. In addition, HYSA treatment increased the expression of osteopontin and the synthesis of osteogenic proteins, such as Type 1 collagen. After the addition of the VDR inhibitor, the effect of HSYA was weakened. CONCLUSION: HSYA could significantly promote the activity and proliferation of osteoblasts and increase the expression level of VDR in osteoblasts. HSYA may also improve calcium absorption by osteoblasts by regulating the synthesis of calciumbinding protein and vitamin D metabolic pathway-related proteins.


Subject(s)
Bone Marrow Cells , Chalcone , Mesenchymal Stem Cells , Osteoblasts , Quinones , Osteoblasts/cytology , Cell Differentiation/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Bone Regeneration/drug effects , Osteoporosis/drug therapy , Cell Proliferation/drug effects , Calcium/metabolism , Receptors, Calcitriol/metabolism , Humans , Chalcone/analogs & derivatives , Chalcone/pharmacology , Quinones/pharmacology
20.
PLoS One ; 17(2): e0262612, 2022.
Article in English | MEDLINE | ID: mdl-35196318

ABSTRACT

Orthodontic treatment requires the regulation of bone remodeling in both compression and tension sides. Transforming growth factor-ß1 (TGF-ß1) is an important coupling factor for bone remodeling. However, the mechanism underlying the TGF-ß1-mediated regulation of the osteoclast-supporting activity of osteoblasts and stromal cells remain unclear. The current study investigated the effect of TGF-ß1 on receptor activator of nuclear factor kappa-B ligand (RANKL) expression in stromal cells induced by 1α,25(OH)2D3 (D3) and dexamethasone (Dex). TGF-ß1 downregulated the expression of RANKL induced by D3 and Dex in mouse bone marrow stromal lineage, ST2 cells. Co-culture system revealed that TGF-ß1 suppressed osteoclast differentiation from bone marrow cell induced by D3 and Dex-activated ST2 cells. The inhibitory effect of TGF-ß1 on RANKL expression was recovered by inhibiting the interaction between TGF-ß1 and the TGF-ß type I/activin receptor or by downregulating of smad2/3 expression. Interestingly, TGF-ß1 degraded the retinoid X receptor (RXR)-α protein which forms a complex with vitamin D receptor (VDR) and regulates transcriptional activity of RANKL without affecting nuclear translocation of VDR and phosphorylation of signal transducer and activator of transcription3 (STAT3). The degradation of RXR-α protein by TGF-ß1 was recovered by a ubiquitin-proteasome inhibitor. We also observed that poly-ubiquitination of RXR-α protein was induced by TGF-ß1 treatment. These results indicated that TGF-ß1 downregulates RANKL expression and the osteoclast-supporting activity of osteoblasts/stromal cells induced by D3 and Dex through the degradation of the RXR-α protein mediated by ubiquitin-proteasome system.


Subject(s)
Osteoclasts/drug effects , Osteoclasts/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology , Ubiquitin/metabolism , Ubiquitination/drug effects , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Coculture Techniques , Leupeptins/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Osteoclasts/cytology , Proteasome Inhibitors/pharmacology , Recombinant Proteins/pharmacology , Signal Transduction/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Transfection , Ubiquitination/genetics
SELECTION OF CITATIONS
SEARCH DETAIL