Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 774
Filter
1.
Am J Physiol Heart Circ Physiol ; 321(5): H920-H932, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34533398

ABSTRACT

Bone marrow-derived endothelial progenitor cells (EPCs) contribute to endothelial repair and angiogenesis. Reduced number of circulating EPCs is associated with future cardiovascular events. We tested whether dysregulated glucose and/or triglyceride (TG) metabolism has an impact on EPC homeostasis. The analysis of metabolic factors associated with circulating EPC number in humans revealed that postprandial hyperglycemia is negatively correlated with circulating EPC number, and this correlation appears to be further enhanced in the presence of postprandial hypertriglyceridemia (hTG). We therefore examined the effect of glucose/TG spikes on bone marrow lineage-sca-1+ c-kit+ (LSK) cells in mice, because primitive EPCs reside in bone marrow LSK fraction. Repetitive glucose + lipid (GL) spikes, but not glucose (G) or lipid (L) spikes alone, induced senescence-like phenotypes of LSK cells, and this phenomenon was reversible after cessation of GL spikes. G spikes and GL spikes differentially affected transcriptional program of LSK cell metabolism and differentiation. GL spikes upregulated a histone H3K27 demethylase JMJD3, and inhibition of JMJD3 eliminated GL spikes-induced LSK cell senescence-like phenotypes. These observations suggest that postprandial glucose/TG dysmetabolism modulate transcriptional regulation in LSK cells through H3K27 demethylase-mediated epigenetic regulation, leading to senescence-like phenotypes of LSK cells, reduced number of circulating EPCs, and development of atherosclerotic cardiovascular disease.NEW & NOTEWORTHY Combination of hyperglycemia and hypertriglyceridemia is associated with increased risk of atherosclerotic cardiovascular disease. We found that 1) hypertriglyceridemia may enhance the negative impact of hyperglycemia on circulating EPC number in humans and 2) metabolic stress induced by glucose + triglyceride spikes in mice results in senescence-like phenotypes of bone marrow stem/progenitor cells via H3K27me3 demethylase-mediated epigenetic regulation. These findings have important implications for understanding the pathogenesis of atherosclerotic cardiovascular disease in patients with T2DM.


Subject(s)
Blood Glucose/metabolism , Bone Marrow Cells/enzymology , Cellular Senescence , DNA Methylation , Diabetes Mellitus, Type 2/blood , Endothelial Progenitor Cells/enzymology , Epigenesis, Genetic , Hyperglycemia/blood , Hypertriglyceridemia/blood , Jumonji Domain-Containing Histone Demethylases/metabolism , Triglycerides/blood , Adult , Aged , Animals , Bone Marrow Cells/pathology , Case-Control Studies , Cell Lineage , Cells, Cultured , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Endothelial Progenitor Cells/pathology , Female , Glycated Hemoglobin , Humans , Hyperglycemia/enzymology , Hyperglycemia/genetics , Hyperglycemia/pathology , Hypertriglyceridemia/enzymology , Hypertriglyceridemia/genetics , Hypertriglyceridemia/pathology , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Mice, Inbred C57BL , Middle Aged , Phenotype
3.
Cell Death Dis ; 12(3): 238, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664230

ABSTRACT

The ubiquitin protease pathway plays important role in human bone marrow-derived mesenchymal stem cell (hBMSC) differentiation, including osteogenesis. However, the function of deubiquitinating enzymes in osteogenic differentiation of hBMSCs remains poorly understood. In this study, we aimed to investigate the role of ubiquitin-specific protease 53 (USP53) in the osteogenic differentiation of hBMSCs. Based on re-analysis of the Gene Expression Omnibus database, USP53 was selected as a positive regulator of osteogenic differentiation in hBMSCs. Overexpression of USP53 by lentivirus enhanced osteogenesis in hBMSCs, whereas knockdown of USP53 by lentivirus inhibited osteogenesis in hBMSCs. In addition, USP53 overexpression increased the level of active ß-catenin and enhanced the osteogenic differentiation of hBMSCs. This effect was reversed by the Wnt/ß-catenin inhibitor DKK1. Mass spectrometry showed that USP53 interacted with F-box only protein 31 (FBXO31) to promote proteasomal degradation of ß-catenin. Inhibition of the osteogenic differentiation of hBMSCs by FBXO31 was partially rescued by USP53 overexpression. Animal studies showed that hBMSCs with USP53 overexpression significantly promoted bone regeneration in mice with calvarial defects. These results suggested that USP53 may be a target for gene therapy for bone regeneration.


Subject(s)
Bone Marrow Cells/enzymology , Mesenchymal Stem Cells/enzymology , Osteogenesis , Ubiquitin-Specific Proteases/metabolism , Adult , Animals , Bone Regeneration , Case-Control Studies , Cells, Cultured , Dependovirus/genetics , F-Box Proteins/metabolism , Genetic Vectors , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mesenchymal Stem Cell Transplantation , Mice, Inbred ICR , Osteoporosis/metabolism , Osteoporosis/pathology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Skull/metabolism , Skull/pathology , Skull/surgery , Tumor Suppressor Proteins/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitination , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism
5.
Mol Med Rep ; 23(2)2021 02.
Article in English | MEDLINE | ID: mdl-33300048

ABSTRACT

Osteoblasts are the main functional cells in bone formation, which are responsible for the synthesis, secretion and mineralization of bone matrix. The PI3K/AKT signaling pathway is strongly associated with the differentiation and survival of osteoblasts. The 3­phosphoinositide­dependent protein kinase­1 (PDK­1) protein is considered the master upstream lipid kinase of the PI3K/AKT cascade. The present study aimed to investigate the role of PDK­1 in the process of mouse osteoblast differentiation in vitro. In the BX­912 group, BX­912, a specific inhibitor of PDK­1, was added to osteoblast induction medium (OBM) to treat bone marrow mesenchymal stem cells (BMSCs), whereas the control group was treated with OBM alone. Homozygote PDK1flox/flox mice were designed and generated, and were used to obtain BMSCsPDK1flox/flox. Subsequently, an adenovirus containing Cre recombinase enzyme (pHBAd­cre­EGFP) was used to disrupt the PDK­1 gene in BMSCsPDK1flox/flox; this served as the pHBAd­cre­EGFP group and the efficiency of the disruption was verified. Western blot analysis demonstrated that the protein expression levels of phosphorylated (p)­PDK1 and p­AKT were gradually increased during the osteoblast differentiation process. Notably, BX­912 treatment and disruption of the PDK­1 gene with pHBAd­cre­EGFP effectively reduced the number of alkaline phosphatase (ALP)­positive cells and the optical density value of ALP activity, as well as the formation of cell mineralization. The mRNA expression levels of PDK­1 in the pHBAd­cre­EGFP group were significantly downregulated compared with those in the empty vector virus group on days 3­7. The mRNA expression levels of the osteoblast­related genes RUNX2, osteocalcin and collagen I were significantly decreased in the BX­912 and pHBAd­cre­EGFP groups on days 7 and 21 compared with those in the control and empty vector virus groups. Overall, the results indicated that BX­912 and disruption of the PDK­1 gene in vitro significantly inhibited the differentiation and maturation of osteoblasts. These experimental results provided an experimental and theoretical basis for the role of PDK­1 in osteoblasts.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases , Bone Marrow Cells/enzymology , Cell Differentiation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Mesenchymal Stem Cells/enzymology , Osteoblasts/enzymology , Protein Kinase Inhibitors/pharmacology , 3-Phosphoinositide-Dependent Protein Kinases/antagonists & inhibitors , 3-Phosphoinositide-Dependent Protein Kinases/biosynthesis , Animals , Male , Mice
7.
J Mol Histol ; 51(1): 67-76, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32002721

ABSTRACT

Age-related jawbone loss directly impact the function of oral cavity resulted from tooth loss, implant failure, and jaw fracture. Numerous evidences show that age-related senescence of bone marrow stromal cells (BMSCs) play a critical role in bone loss, but little attention has been paid to jawbone. Here, we delineated the critical role of sirtuin family protein 6 (SIRT6) in senescence, autophagy, and osteogenesis of BMSCs from jawbones. Radiography analysis showed less jawbone quality in elderly than young people. We also showed that SIRT6 expression decreased in bone tissue and BMSCs from the elderly by immunochemical staining. BMSCs from the elderly exhibited decreased osteogenic differentiation and inclined senescence which these phenotypes could be simulated by SIRT6 knockdown. Furthermore, accompanied with the inhibition of SIRT6, the autophagy level and ostogenesis of BMSCs was also decreased. However, using rapamycin, an autophagy activator, could rescue these adverse effects of BMSCs caused by SIRT6 inhibition. Mechanistically, SIRT6 regulated the autophagy and osteogenesis of BMSCs by activating AKT-mTOR pathway, at least in part. Finally, a decreased jawbone quality was shown in SIRT6 haploinsufficiency mice by Wnt1 specific tissue knockdown (Wnt1-Cre;SIRT6fl/+) model. Taken together, our data revealed that SIRT6 adjusted senescence and osteogenesis of BMSCs via altering autophagy level, and associated with age-related bone loss. SIRT6 could be as a promising therapeutic target for age-related osteoporosis of jawbone.


Subject(s)
Aging/metabolism , Bone Marrow Cells/enzymology , Jaw/enzymology , Mesenchymal Stem Cells/enzymology , Sirtuins/metabolism , Adult , Aged , Aging/genetics , Animals , Bone Marrow Cells/cytology , Humans , Jaw/cytology , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Knockout , Middle Aged , Osteogenesis/genetics , Sirtuins/genetics
8.
J Orthop Sci ; 25(2): 331-337, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31072650

ABSTRACT

BACKGROUND: Mechanical unloading induces bone loss in human weight-loaded bones. The findings of recent studies have revealed that cluster of differentiation 38 knockout mice display bone loss similar to that observed in osteoporosis. This study aimed to determine whether the expression of cluster of differentiation 38 is implicated in skeletal unloading and reloading. METHODS: Eight-week-old male C57BL/6J mice were assigned to control, tail-suspension, or reloading after tail-suspension groups. In the tail-suspension group, tail suspension elevated the hind limbs for 1 week. The bilateral femurs and tibias from the groups were evaluated for cluster of differentiation 38 immunocytochemistry, and the cluster of differentiation 38 messenger ribonucleic acid levels and the expression of cluster of differentiation 38 and other cell-surface antigens were evaluated using quantitative real-time polymerase chain reaction and flow cytometric analyses. RESULTS: In the tail-suspension group, the alkaline phosphatase reactivity, cluster of differentiation 38 immunoreactivity in the bone marrow and osteoblasts, and the expression of cluster of differentiation 38 messenger ribonucleic acid and that of other cell-surface antigens were significantly lower than those in the control group. In the reloading after tail-suspension group, the level of cluster of differentiation 38 expression was restored to the same level as that in the control group. CONCLUSIONS: Cluster of differentiation 38 expression declined after skeletal unloading and recovered to normal levels after reloading. In the bone marrow, cluster of differentiation 38 expression plays a crucial role in bone formation in response to mechanical stress.


Subject(s)
ADP-ribosyl Cyclase 1/physiology , Bone Diseases, Metabolic/enzymology , Bone Marrow Cells/enzymology , Cyclic ADP-Ribose/metabolism , Osteoblasts/enzymology , Weight-Bearing , Animals , Femur/metabolism , Hindlimb Suspension , Male , Mice , Mice, Inbred C57BL
9.
Stem Cell Res Ther ; 10(1): 351, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31775910

ABSTRACT

BACKGROUND: Silicon-modified biomaterials have been extensively studied in bone tissue engineering. In recent years, the toxicity of silicon-doped biomaterials has gradually attracted attention but requires further elucidation. This study was designed to explore whether high-dose silicate can induce a cytotoxicity effect in bone mesenchymal stem cells (BMSCs) and the role of autophagy in its cytotoxicity and mechanism. METHODS: Morphologic changes and cell viability of BMSCs were detected after different doses of silicate exposure. Autophagic proteins (LC3, p62), LC3 turnover assay, and RFP-GFP-LC3 assay were applied to detect the changes of autophagic flux following silicate treatment. Furthermore, to identify the potential mechanism of autophagic dysfunction, we tested the acetyl-α-tubulin protein level and histone deacetylase 6 (HDAC6) activity after high-dose silicate exposure as well as the changes in microtubule and autophagic activity after HDAC6 siRNA was applied. RESULTS: It was found that a high dose of silicate could induce a decrease in cell viability; LC3-II and p62 simultaneously increased after high-dose silicate exposure. A high concentration of silicate could induce autophagic dysfunction and cause autophagosomes to accumulate via microtubule destabilization. Results showed that acetyl-α-tubulin decreased significantly with high-dose silicate treatment, and inhibition of HDAC6 activity can restore microtubule structure and autophagic flux. CONCLUSIONS: Microtubule destabilization caused by a high concentration of silicate via HDAC6 activation contributed to autophagic dysfunction in BMSCs, and inhibition of HDAC6 exerted a cytoprotection effect through restoration of the microtubule structure and autophagic flux.


Subject(s)
Autophagic Cell Death/drug effects , Bone Marrow Cells/enzymology , Histone Deacetylase 6/metabolism , Mesenchymal Stem Cells/enzymology , Microtubules/metabolism , Silicates/pharmacology , Animals , Bone Marrow Cells/cytology , Enzyme Activation/drug effects , Mesenchymal Stem Cells/cytology , Rats , Silicates/adverse effects
10.
Cancer Lett ; 459: 100-111, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31158430

ABSTRACT

The mixed lineage kinase domain-like protein (MLKL) has emerged as a critical mediator of necroptosis, which results in the release of cellular damage-associated molecular patterns (DAMPs). However, its physiological role in regulating inflammation is not fully understood. We herein showed that Mlkl-/- mice were highly susceptible to colitis and colitis-associated tumorigenesis (CAT), which was associated with massive leukocyte infiltration and increased inflammatory responses. Moreover, we used bone marrow transplantation to reveal that MLKL in inflammatory cells is crucial for its role on colitis. Intestinal mucosal tissue and polyps isolated from Mlkl-/- mice exhibited increased ERK activation and elevated expression of genes associated with inflammation and cancer. Mechanistically, enhanced inflammation in Mlkl-/- mice was due to MEK/ERK activation particularly in dendritic cells (DCs). Our results demonstrate the role of MLKL in maintaining intestinal homeostasis and protecting against colitis and tumorigenesis.


Subject(s)
Colitis/enzymology , Colonic Neoplasms/immunology , Protein Kinases/immunology , Animals , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Colitis/genetics , Colitis/immunology , Colitis/pathology , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cytokines/biosynthesis , Dendritic Cells/enzymology , Dendritic Cells/immunology , Dendritic Cells/pathology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Intestinal Mucosa/enzymology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Macrophages/enzymology , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Kinases/deficiency , Protein Kinases/genetics
11.
Br J Haematol ; 185(5): 883-887, 2019 06.
Article in English | MEDLINE | ID: mdl-30873593

ABSTRACT

Within lymphoid tissues, chronic lymphocytic leukaemia (CLL) cells interact with mesenchymal stromal cells (MSC). Inhibitors of phosphoinositide 3-kinase delta (PI3Kδ) cause release of CLL cells from lymphoid tissues into blood. PI3Kδ inhibitors are thought to target only CLL and other immune cells because PI3Kδ expression is restricted to haematopoietic cells. We found that PI3Kδ is unexpectedly expressed in primary MSC derived from CLL patients and healthy donors. PI3Kδ inhibition in MSC using idelalisib or duvelisib significantly reduced their ability to support CLL migration and adhesion. These observations provide the first evidence that PI3Kδ is expressed and functional in CLL MSC.


Subject(s)
Bone Marrow Cells/enzymology , Class I Phosphatidylinositol 3-Kinases/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Mesenchymal Stem Cells/enzymology , Antineoplastic Agents/pharmacology , Bone Marrow Cells/pathology , Case-Control Studies , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/biosynthesis , Class I Phosphatidylinositol 3-Kinases/genetics , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Isoquinolines/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mesenchymal Stem Cells/pathology , Purines/pharmacology , Quinazolinones/pharmacology
12.
Blood Adv ; 3(6): 908-916, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30898762

ABSTRACT

An intriguing aspect of the clinical activity of FMS-like tyrosine kinase 3 inhibitors (FLT3 TKIs) is their apparent higher activity against peripheral blasts from FLT3/internal tandem duplication (ITD) acute myeloid leukemia than marrow disease in the same patients. Accordingly, studies showed that the bone marrow microenvironment plays a role in FLT3 TKI resistance, although the underlying mechanisms are unclear. We recently identified a previously undescribed mechanism by which the bone marrow microenvironment can contribute to drug resistance: expression of cytochrome P450 enzymes (CYPs). In fact, bone marrow stromal cells (BMSCs) expressed most CYPs, including CYP3A4. Because hepatic CYP3A4 plays a role in the inactivation of several FLT3 TKIs, we explored the potential role of CYP3A4 in bone marrow microenvironment-mediated FLT3 TKI resistance. We found that CYP3A4 plays a major role in BMSC-mediated inhibition in the activity of 3 different FLT3 TKIs (sorafenib, quizartinib, and gilteritinib) against FLT3/ITD acute myeloid leukemia (AML). Furthermore, clarithromycin, a clinically active CYP3A4 inhibitor, significantly reversed the protective effects of BMSCs. We show, for the first time, that bone marrow stromal CYP3A4 contributes to FLT3 TKI resistance in the bone marrow. These results suggest that combining FLT3 TKIs with CYP3A4 inhibitors could be a promising strategy toward improving the activity of FLT3 TKIs.


Subject(s)
Cytochrome P-450 CYP3A/physiology , Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Bone Marrow Cells/enzymology , Cells, Cultured , Cytochrome P-450 CYP3A/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Protein Kinase Inhibitors/metabolism , Stromal Cells/enzymology , Stromal Cells/metabolism , Tandem Repeat Sequences , Tumor Microenvironment , fms-Like Tyrosine Kinase 3/genetics
13.
Iran Biomed J ; 23(2): 142-52, 2019 03.
Article in English | MEDLINE | ID: mdl-30376703

ABSTRACT

Background: Since the low concentration and short-time treatment with sodium nitroprusside (SNP), a nitric oxide (NO)­donor, cause no harm to rat bone marrow mesenchymal stem cells (MSCs), we studied the impact of SNP on MSCs differentiation. Methods: MSCs were treated with 100 and 1000 µM of SNP for 1 hour in every 48 hours and after 5, 10, 15, and 21 days in osteogenic media. The viability and the level of mineralization were determined using MTT assay and alizarin red staining, respectively. Morphology of the cells was studied using fluorescent dye. Concentration of calcium and the activity of alanine transaminase (ALT), aspartate transaminase (AST), lactate dehydrogenase (LDH), and alkaline phosphatase (ALP) were evaluated by commercial kits. Results: SNP with the concentration of 1000 µM significantly reduced viability from day 5 to day 20, but 100 µM did not affect the viability until the day 15. The low concentration of SNP increased matrix deposition from day 10 and reached almost its maximum (4.40 ± 2.4) at the day 15. Also, increasing the activity of ALP (419 ± 2.2), due to low concentration of SNP, started at day 10 and continued till the day 20, while LDH (2026 ± 11) and AST (25.6 ± 0.4) elevations were observed from day 5 onwards. In case of ALT, we observed a significant decrease (36%) from day 5 till day 20. Conclusion: Based on our findings, low concentrations of SNP might be useful in the promotion of bone repair.


Subject(s)
Alkaline Phosphatase/metabolism , Bone Density/drug effects , Bone Marrow Cells/drug effects , Mesenchymal Stem Cells/drug effects , Nitric Oxide/pharmacology , Animals , Bone Density/physiology , Bone Marrow Cells/enzymology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Mesenchymal Stem Cells/enzymology , Nitroprusside/pharmacology , Rats , Rats, Wistar
14.
J Thromb Haemost ; 17(1): 19-30, 2019 01.
Article in English | MEDLINE | ID: mdl-30489000

ABSTRACT

Essentials Plasma Factor XIII, a heterodimer of A and B subunits FXIIIA2 B2 , is a transglutaminase enzyme with a well-established role in haemostasis. Cells of bone marrow and mesenchymal lineage express the FXIII-A gene (F13A1) that encodes the cellular form of the transglutaminase, a homodimer of the A subunits, FXIII-A. FXIII-A was presumed to function intracellularly, however, several lines of evidence now indicate that FXIII-A is externalised by an as yet unknown mechanism This review describes the mounting evidence that FXIII-A is a diverse transglutaminase with many intracellular and extracellular substrates that can participate in an array of biological processes SUMMARY: Factor XIII is a tranglutaminase enzyme that catalyzes the formation of ε-(γ-glutamyl)lysyl isopeptide bonds in protein substrates. The plasma form, FXIII-A2 B2 , has an established function in hemostasis, where its primary substrate is fibrin. A deficiency in FXIII manifests as a severe bleeding diathesis, underscoring its importance in this pathway. The cellular form of the enzyme, a homodimer of the A-subunits, denoted FXIII-A, has not been studied in as extensive detail. FXIII-A was generally perceived to remain intracellular, owing to the lack of a classical signal peptide for its release. In the last decade, emerging evidence has revealed that this diverse transglutaminase can be externalized from cells, by an as yet unknown mechanism, and can cross-link extracellular substrates and participate in a number of diverse pathways. The FXIII-A gene (F13A1) is expressed in cells of bone marrow and mesenchymal lineage, notably megakaryocytes, monocytes/macrophages, dendritic cells, chrondrocytes, osteoblasts, and preadipocytes. The biological processes that FXIII-A is coupled with, such as wound healing, phagocytosis, and bone and matrix remodeling, reflect its expression in these cell types. This review describes the mounting evidence that this cellular transglutaminase can be externalized, usually in response to stimuli, and participate in extracellular cross-linking reactions. A corollary of being involved in these biological pathways is the participation of FXIII-A in pathological processes. In conclusion, the functions of this transglutaminase extend far beyond its role in hemostasis, and our understanding of this enzyme in terms of its secretion, regulation and substrates is in its infancy.


Subject(s)
Bone Marrow Cells/enzymology , Factor XIIIa/metabolism , Hemostasis , Mesenchymal Stem Cells/enzymology , Animals , Cell Lineage , Factor XIIIa/genetics , Humans , Protein Multimerization , Protein Structure, Quaternary , Protein Transport , Signal Transduction , Substrate Specificity
15.
Immunol Lett ; 205: 40-50, 2019 01.
Article in English | MEDLINE | ID: mdl-30447309

ABSTRACT

Human myeloma cells grow in a hypoxic acidic niche in the bone marrow. Cross talk among cellular components of this closed niche generates extracellular adenosine, which promotes tumor cell survival. This is achieved through the binding of adenosine to purinergic receptors into complexes that function as an autocrine/paracrine signal factor with immune regulatory activities that i) down-regulate the functions of most immune effector cells and ii) enhance the activity of cells that suppress anti-tumor immune responses, thus facilitating the escape of malignant myeloma cells from immune surveillance. Here we review recent findings confirming that the dominant phenotype for survival of tumor cells is that where the malignant cells have been metabolically reprogrammed for the generation of lactic acidosis in the bone marrow niche. Adenosine triphosphate and nicotinamide-adenine dinucleotide extruded from tumor cells, along with cyclic adenosine monophosphate, are the main intracellular energetic/messenger molecules that serve as leading substrates in the extracellular space for membrane-bound ectonucleotidases metabolizing purine nucleotides to signaling adenosine. Within this mechanistic framework, the adenosinergic substrate conversion can vary significantly according to the metabolic environment. Indeed, the neoplastic expansion of plasma cells exploits both enzymatic networks and hypoxic acidic conditions for migrating and homing to a protected niche and for evading the immune response. The expression of multiple specific adenosine receptors in the niche completes the profile of a complex regulatory framework whose signals modify multiple myeloma and host immune responses.


Subject(s)
Adenine Nucleotides/metabolism , Adenosine/metabolism , Antigens, CD/metabolism , Bone Marrow Cells/enzymology , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Bone Marrow Cells/immunology , Humans , Immunosuppression Therapy , Multiple Myeloma/immunology , Receptors, Purinergic/metabolism , Signal Transduction/immunology , Tumor Microenvironment/immunology
16.
Methods Mol Biol ; 1821: 219-233, 2018.
Article in English | MEDLINE | ID: mdl-30062415

ABSTRACT

The actin cytoskeleton is essential for the biology of osteoclasts, in particular during bone resorption. As key regulators of actin dynamics, the small GTPases of the Rho family are very important in the control of osteoclast activity. The study of Rho GTPase signaling pathways is essential to uncover the mechanisms of bone resorption and can have interesting applications for the treatment of osteolytic diseases. In this chapter, we describe various techniques to obtain primary osteoclasts from murine bone marrow cells, to measure Rho GTPase activation levels, to monitor bone resorption activity of osteoclasts and to introduce the expression of proteins of interest using a retroviral approach. We illustrate the different methods with experimental examples of the effect of Rac1 activation by the exchange factor Dock5 on bone resorption by osteoclasts.


Subject(s)
Bone Marrow Cells/enzymology , Bone Resorption/enzymology , Neuropeptides/metabolism , Osteoclasts/enzymology , Signal Transduction , rac1 GTP-Binding Protein/metabolism , Animals , Bone Marrow Cells/pathology , Bone Resorption/pathology , Cells, Cultured , Guanine Nucleotide Exchange Factors/metabolism , Mice , Osteoclasts/pathology
17.
Biomed Pharmacother ; 106: 1126-1134, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30119179

ABSTRACT

Senescence has become a hot point issue in recent decades and requires urgent attention. As a novel and effective antioxidant, hydrogen has been proved to alleviate cellular senescence in endothelial cells in vitro. However, the effects and mechanisms of hydrogen on senescence in vivo are still unclear. In the present study, 12-month-old Sprague Dawley (SD) rats were intraperitoneal administration of hydrogen-rich saline (HRS, 10 ml/kg). Subsequently, bone marrow-derived stem cells (BMSCs) were harvested for the detection of hydrogen antisenescence effects and mechanisms. The results showed that the number of senescence-associated ß-galactosidase (SA-ß-Gal) positive cells was reduced in BMSCs from rats treated with HRS. BMSCs in rats treated with HRS possessed a better proliferation ability, showed more effectively tri-lineage differentiation potential, and had less percentage of cells in G1 cell cycle arrest than the control cells. Additionally, HRS administration inhibited the production of intracellular reactive oxygen species (ROS) and decreased the expression of senescence-related proteins p53 and p21. Our results revealed that hydrogen could alleviate cellular senescence in vivo. And the underlying mechanism of antisenescence effects of hydrogen in BMSCs was via the ROS/p53/p21 signaling pathway. Thus, hydrogen could be a new and convenient strategy for alleviating senescence and for therapy of age-related diseases.


Subject(s)
Antioxidants/pharmacology , Bone Marrow Cells/drug effects , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Hydrogen/pharmacology , Mesenchymal Stem Cells/drug effects , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Age Factors , Animals , Bone Marrow Cells/enzymology , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured , Dose-Response Relationship, Drug , G1 Phase Cell Cycle Checkpoints/drug effects , Male , Mesenchymal Stem Cells/enzymology , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors
18.
Food Chem Toxicol ; 120: 243-252, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29964085

ABSTRACT

A majority of cellular diseases, independent of their origin, are characterized by a dramatic increase in Reactive Oxygen Species (ROS) in response to stress. In most cases, the uncontrolled detrimental ROS outburst is difficult to handle for the cellular machinery and eventually leads to cell mortality. In this study, we compare the antioxidant efficacy of quercetin and melatonin to find out a better alternative against lipopolysaccharide (LPS) induced tissue injury by oxidative stress in Funambulus pennanti. Transient exposure to LPS significantly increased ROS generation and lipid peroxidation levels in bone marrow mononuclear cells (MNCs) and spleen which was further corroborated by decreased activities of SOD, CAT and Gpx enzymes. It also downregulate the expression of cellular oxidative stress response proteins Nrf-2 and HO-1 in spleen and decreases the proliferation of bone marrow derived Granulocyte macrophage-colony forming unit cells (GM-CFU). Both melatonin and quercetin pre-treatments rescued these effects, however, our results indicated that the efficacy of melatonin to overcome oxidative stress was significantly better than quercetin. Our findings support the idea that melatonin is a better antioxidant and immunomodulator as compared to other alternatives and perhaps may be employed in the development of effective therapeutics against ROS dominated diseases.


Subject(s)
Bone Marrow Cells/drug effects , Lipopolysaccharides/toxicity , Melatonin/pharmacology , Oxidative Stress/drug effects , Quercetin/pharmacology , Spleen/drug effects , Animals , Biomarkers/metabolism , Bone Marrow Cells/enzymology , Bone Marrow Cells/metabolism , Catalase/metabolism , Glutathione Peroxidase/metabolism , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Sciuridae , Spleen/cytology , Spleen/enzymology , Spleen/metabolism , Superoxide Dismutase/metabolism
19.
J Biol Chem ; 293(33): 12934-12944, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29907570

ABSTRACT

Humoral immunity involves multiple checkpoints that occur in B cell development, maturation, and activation. The pre-B-cell receptor (pre-BCR) is expressed following the productive recombination of the immunoglobulin heavy-chain gene, and sSignalsing through the pre-BCR are required for the differentiation of pre-B cells into immature B cells. However, the molecular mechanisms controlling the pre-BCR expression and signaling strength remain undefined. Herein, we probed the role of the endoplasmic reticulum-associated, stress-activated E3 ubiquitin ligase HMG-CoA reductase degradation 1 (Hrd1) in B cell differentiation. Using mice with a specific Hrd1 deletion in pro-B cells and subsequent B cell developmental stages, we showed that the E3 ubiquitin ligase Hrd1 governs a critical checkpoint during B cell development. We observed that Hrd1 is required for degradation of the pre-BCR complex during the early stage of B cell development. As a consequence, loss of Hrd1 in the B cell lineage resulted in increased pre-BCR expression levels and a developmental defect in the transition from large to small pre-B cells. This defect, in turn, resulted in reduced fewer mature B cells in bone marrow and peripheral lymphoid organs. Our results revealed a novel critical role of Hrd1 in controlling a critical checkpoint in B cell-mediated immunity and suggest that Hrd1 may functioning as an E3 ubiquitin ligase of the pre-BCR complex.


Subject(s)
Bone Marrow Cells/immunology , Cell Differentiation/immunology , Endoplasmic Reticulum/immunology , Precursor Cells, B-Lymphoid/immunology , Receptors, Antigen, B-Cell/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Cell Differentiation/genetics , Cell Line , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/genetics , Mice , Mice, Transgenic , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/enzymology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
20.
Basic Res Cardiol ; 113(3): 18, 2018 03 17.
Article in English | MEDLINE | ID: mdl-29549541

ABSTRACT

In addition to being a peptidase, the angiotensin-converting enzyme (ACE) can be phosphorylated and involved in signal transduction. We evaluated the role of ACE in granulocyte-colony-stimulating factor (G-CSF)-induced hematopoietic progenitor cell (HPC) mobilization and detected a significant increase in mice-lacking ACE. Transplantation experiments revealed that the loss of ACE in the HPC microenvironment rather than in the HPCs increased mobilization. Indeed, although ACE was expressed by a small population of bone-marrow cells, it was more strongly expressed by endosteal bone. Interestingly, there was a physical association of ACE with the G-CSF receptor (CD114), and G-CSF elicited ACE phosphorylation on Ser1270 in vivo and in vitro. A transgenic mouse expressing a non-phosphorylatable ACE (ACES/A) mutant demonstrated increased G-CSF-induced HPC mobilization and decreased G-CSF-induced phosphorylation of STAT3 and STAT5. These results indicate that ACE expression/phosphorylation in the bone-marrow niche interface negatively regulates G-CSF-induced signaling and HPC mobilization.


Subject(s)
Bone Marrow Cells/drug effects , Bone Marrow/drug effects , Bone and Bones/drug effects , Cell Movement/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Peptidyl-Dipeptidase A/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Bone Marrow/enzymology , Bone Marrow Cells/enzymology , Bone and Bones/enzymology , Cell Proliferation/drug effects , Hematopoietic Stem Cells/enzymology , Mice, Inbred C57BL , Mice, Knockout , Peptidyl-Dipeptidase A/deficiency , Peptidyl-Dipeptidase A/genetics , Phosphorylation , Ramipril/pharmacology , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Stem Cell Niche
SELECTION OF CITATIONS
SEARCH DETAIL
...