Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 328
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Mol Biol Rep ; 51(1): 261, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38302805

ABSTRACT

BACKGROUND: The cardioprotective properties of mesenchymal stem cells and the therapeutic potential of curcumin (CUR) have been explored. Combining these approaches may enhance stem cell effectiveness and expedite healing. This study aimed to investigate the synergistic effects of co-treating bone marrow mesenchymal stem cells (BMSCs) with curcumin on vascular endothelial growth factor (VEGF) levels, in a rat model of myocardial ischemia (MI). METHODS AND RESULTS: Sixty-five male rats were divided into four groups: G1 (healthy control), G2 (MI induced by isoproterenol hydrochloride), G3 (treated with BMSCs), and G4 (co-treated with curcumin and BMSCs). Blood and tissue samples were collected at specific time points (day 1, 7, 15 and 21) after MI induction. Serum levels of lactate dehydrogenase (LDH), creatine kinase (CK), cardiac troponin I (cTnI), aspartate aminotransferase (AST), CK-MB and VEGF were measured. VEGF mRNA and protein expression were evaluated using RT-qPCR and Western blot techniques. Histopathological assessments were performed using H&E staining and CD31 immunofluorescence staining. VEGF expression significantly increased on days 7 and 15 in the CUR-BMSCs group, peaking on day 7. Western blot analysis confirmed elevated VEGF protein expression on days 7 and 15 post-MI. ELISA results demonstrated increased serum VEGF levels on days 7 and 15, reaching the highest level on day 7 in CUR-BMSCs-treated animals. Treated groups showed lower levels of LDH, AST, CK, CK-MB and cTnI compared to the untreated MI group. H&E staining revealed improved myocardial structure, increased formation of new capillaries, in both treatment groups compared to the MI group. CONCLUSION: Combining curcumin with BMSCs promotes angiogenesis in the infarcted myocardium after 15 days of MI induction. These findings suggest the potential of this combined therapy approach for enhancing cardiac healing and recovery.


Subject(s)
Coronary Artery Disease , Curcumin , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Myocardial Infarction , Myocardial Ischemia , Rats , Male , Animals , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Curcumin/pharmacology , Curcumin/metabolism , Bone Marrow/metabolism , Angiogenesis , Myocardial Ischemia/metabolism , Myocardium/metabolism , Coronary Artery Disease/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cell Transplantation/methods , Bone Marrow Cells
2.
Biochem Biophys Res Commun ; 682: 132-137, 2023 11 19.
Article in English | MEDLINE | ID: mdl-37806251

ABSTRACT

Bone marrow derived cells (BMDCs) migrate into the hypothalamus, where those cells give rise to microglia to regulate food intake. Given the fact that diabetes functionally impairs BMDCs, we hypothesized that diabetic microglia would fail to exhibit physiological function, accounting for hyperphagia in diabetes. To examine the role of BMDCs, total bone marrow cells from GFP transgenic mice were transplanted into wild type mice in which diabetes was induced by streptozotocin. We first confirmed that bone marrow transplantation could be utilized to examine BMDCs in the brain parenchyma as GFP positive cells could engraft the brain parenchyma and give rise to microglia even when the BBB was intact in the recipient mice. While diabetic mice manifested hyperphagia, BMDCs were in smaller number in the hypothalamus with less response to fasting in the brain parenchyma compared to nondiabetic mice. This finding was also confirmed by examining nondiabetic chimera mice in which BMDCs were diabetic. Those mice also exhibited less response of BMDCs in response to fasting. In conclusion, diabetic BMDCs had less response of microglia to fasting, perhaps accounting for diabetic hyperphagia.


Subject(s)
Bone Marrow , Diabetes Mellitus, Experimental , Mice , Animals , Bone Marrow/metabolism , Microglia/metabolism , Appetite , Mice, Transgenic , Bone Marrow Transplantation , Bone Marrow Cells/metabolism , Hyperphagia , Hypothalamus/metabolism , Mice, Inbred C57BL , Green Fluorescent Proteins/metabolism
3.
Commun Biol ; 6(1): 1043, 2023 10 14.
Article in English | MEDLINE | ID: mdl-37833362

ABSTRACT

Obesity adversely affects bone and fat metabolism in mice and humans. Omega-3 polyunsaturated fatty acids (omega-3 PUFAs) have been shown to improve glucose metabolism and bone homeostasis in obesity. However, the impact of omega-3 PUFAs on bone marrow adipose tissue (BMAT) and bone marrow stromal cell (BMSC) metabolism has not been intensively studied yet. In the present study we demonstrated that omega-3 PUFA supplementation in high fat diet (HFD + F) improved bone parameters, mechanical properties along with decreased BMAT in obese mice when compared to the HFD group. Primary BMSCs isolated from HFD + F mice showed decreased adipocyte and higher osteoblast differentiation with lower senescent phenotype along with decreased osteoclast formation suggesting improved bone marrow microenvironment promoting bone formation in mice. Thus, our study highlights the beneficial effects of omega-3 PUFA-enriched diet on bone and cellular metabolism and its potential use in the treatment of metabolic bone diseases.


Subject(s)
Bone Marrow , Fatty Acids, Omega-3 , Humans , Mice , Animals , Bone Marrow/metabolism , Adiposity , Bone and Bones/metabolism , Obesity/complications , Obesity/prevention & control , Obesity/metabolism , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/metabolism , Disease Models, Animal
4.
Int J Mol Sci ; 24(10)2023 May 16.
Article in English | MEDLINE | ID: mdl-37240169

ABSTRACT

During aging, bone marrow mesenchymal stromal cells (MSCs)-the precursors of osteoblasts-undergo cellular senescence, losing their osteogenic potential and acquiring a pro-inflammatory secretory phenotype. These dysfunctions cause bone loss and lead to osteoporosis. Prevention and intervention at an early stage of bone loss are important, and naturally active compounds could represent a valid help in addition to diet. Here, we tested the hypothesis that the combination of two pro-osteogenic factors, namely orthosilicic acid (OA) and vitamin K2 (VK2), and three other anti-inflammatory compounds, namely curcumin (CUR), polydatin (PD) and quercetin (QCT)-that mirror the nutraceutical BlastiMin Complex® (Mivell, Italy)-would be effective in promoting MSC osteogenesis, even of replicative senescent cells (sMSCs), and inhibiting their pro-inflammatory phenotype in vitro. Results showed that when used at non-cytotoxic doses, (i) the association of OA and VK2 promoted MSC differentiation into osteoblasts, even when cultured without other pro-differentiating factors; and (ii) CUR, PD and QCT exerted an anti-inflammatory effect on sMSCs, and also synergized with OA and VK2 in promoting the expression of the pivotal osteogenic marker ALP in these cells. Overall, these data suggest a potential role of using a combination of all of these natural compounds as a supplement to prevent or control the progression of age-related osteoporosis.


Subject(s)
Bone Diseases, Metabolic , Curcumin , Mesenchymal Stem Cells , Osteoporosis , Humans , Osteogenesis , Quercetin/therapeutic use , Vitamin K 2/pharmacology , Vitamin K 2/metabolism , Curcumin/pharmacology , Bone Marrow/metabolism , Cell Differentiation , Mesenchymal Stem Cells/metabolism , Osteoporosis/drug therapy , Osteoporosis/metabolism , Bone Diseases, Metabolic/metabolism , Cells, Cultured , Bone Marrow Cells
5.
Phytother Res ; 37(1): 89-100, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36161389

ABSTRACT

Inflammatory bowel disease is a disease that can invade the whole digestive tract and is accompanied by immune abnormalities. Immune dysfunction involving dendritic cells (DCs) and T cells is recognized as a key factor in diseases. Indirubin (IDRB) exerts antiinflammatory effects and can help in treating immune diseases. This study aimed to isolate bone marrow-derived dendritic cells (BMDCs) using lipopolysaccharide (LPS) to obtain mature DCs (mDCs). The expression of CD80, CD86, CD40, and MHC-II was detected using flow cytometry after treatment with IDRB. αVß8 siRNA was used to knock down αVß8 in mDCs, and the expression of CD80, CD86, CD40, and MHC-II was detected. Meanwhile, DCs were co-cultured with T cells. Then, T cell differentiation was detected using flow cytometry, and the cytokine levels were detected using enzyme-linked immunosorbent assay. The animal model of dextran sulfate sodium (DSS)-induced inflammatory bowel disease was established in mice. After intervention with IDRB and αVß8 shRNA, the intestinal tissues were evaluated using H&E staining, disease activity index (DAI) score, and histological damage index, and the corresponding factors and cytokines to regulatory T cells (Treg) and Th17 were measured. The results showed that αVß8 was expressed in immature DCs and mDCs. CD80, CD86, CD40, and MHC-II expression decreased after IDRB treatment in mDCs. Meanwhile, the expression of TNF-α and TGF-ß also decreased after IDRB treatment. The effect of IDRB on the expression of CD80, CD86, CD40, MHC-II, TNF-α, and TGF-ß in mDCs was reversed by αVß8 siRNA. The Treg differentiation increased after IDRB treatment, while the differentiation of Th17 cells was inhibited. This effect of IDRB was reversed by mDCs after treatment with αVß8 siRNA. In vivo experiments showed that IDRB alleviated the symptoms of inflammatory bowel disease in animals. Enteritis significantly reduced, and the effect of IDRB was reversed by αVß8 shRNA. The results suggested that IDRB regulated the differentiation of T cells by mediating the maturation of BMDCs through αVß8. This study confirmed the therapeutic effect of IDRB in inflammatory bowel disease and suggested that IDRB might serve as a potential drug.


Subject(s)
Inflammatory Bowel Diseases , Tumor Necrosis Factor-alpha , Mice , Animals , Tumor Necrosis Factor-alpha/metabolism , Bone Marrow/metabolism , Cell Differentiation , Cytokines/metabolism , Transforming Growth Factor beta/pharmacology , Cells, Cultured , Inflammatory Bowel Diseases/drug therapy , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Dendritic Cells/metabolism , Mice, Inbred C57BL
6.
Gene ; 849: 146902, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36169052

ABSTRACT

Different studies indicated that the enhancing the expression of germ cell markers improved the efficiency of stem cells in the generation of germ line cells. The aim of the present study was to investigate the effect of SAG-dihydrochloride on the expression of germ cell markers in the human bone marrow-mesenchymal stem cells (BM-MSCs). For this purpose, the human BM-MSCs were cultured in the medium containing different concentrations of SAG-dihydrochloride (10, 20 and 30 µM). After RNA extraction and cDNA synthesis, the expression level of PTCH1, GLI1, PLZF, DDX4 and STRA8 genes were determined by using SYBR Green Real time PCR. The analysis of the results obtained from PTCH1 and GLI1 expression indicated that SAG-dihydrochloride had the ability to enhance the expression of germ cell markers in a Gli-independent manner. Furthermore, the significant increased expression of STRA8 was observed in the BM-MSCs treated by 10 µM SAG-dihydrochloride for 4 and 6 days (p < 0.05). There was also the up-regulation of DDX4 in the BM-MSCs following treatment with 20 µM SAG-dihydrochloride for 4 and 6 days. The obtained results suggested that treatment with SAG-dihydrochloride increased the expression of germ cell markers in the human BM-MSCs through the activation of non-canonical sonic hedgehog signaling pathway.


Subject(s)
Bone Marrow Cells , Mesenchymal Stem Cells , Humans , Bone Marrow Cells/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Cell Differentiation/genetics , DNA, Complementary , Bone Marrow/metabolism , Zinc Finger Protein GLI1/genetics , Mesenchymal Stem Cells/metabolism , Germ Cells/metabolism , RNA
7.
Toxicology ; 481: 153354, 2022 11.
Article in English | MEDLINE | ID: mdl-36265525

ABSTRACT

Melatonin (MLT) was reported to have therapeutic effects on inflammatory bowel disease (IBD) such as ulcerative colitis (UC) and Crohn's disease (CD) due to its anti-inflammatory and immunomodulatory properties. However, whether the beneficial effects of melatonin on colitis are through altering the immune response of bone marrow-derived dendritic cells (BMDCs) has not been well characterized. Here, we propose that MLT alleviates dextran sulfate sodium (DSS)-induced colitis in mice through its regulation of the immune response of BMDCs, in which some lncRNA, circRNA, miRNA, and mRNA may be involved. We at first established a DSS-induced colitis mouse model and found that the concentration of MLT in the serum of DSS-induced colitis mice was significantly lower than that in the control mice. Supplementation with MLT alleviated DSS-induced colitis in mice, which was reflected by preventing mouse body weight loss, colon length shortening, inflammation, and epithelial tissue destruction and abscission in the colon. We then isolated and cultured BMDCs and found that MLT could inhibit the activation of BMDCs from the colitis mice, which was reflected by reducing the phagocytotic ability of the cells, inhibiting their migration, and decreasing their secretion of pro-inflammatory cytokines. RNA sequencing results showed that MLT promoted the transformation of BMDCs into immune tolerant phenotypes in DSS-induced colitis mice through affecting non-coding RNAs (ncRNAs). Among them, lncRNA ENSMUST00000226323, circRNA-0520, and circRNA-2243 were predicted to interact with miRNA-709, and mRNAs of Ywhaz and Ccl9 were the targets of miRNA-709, all of which were involved in MLT-induced alteration of BMDCs functions in DSS-induced colitis mice via PI3K-Akt pathway. Our findings may provide some clues for understanding MLT inhibiting inflammatory response in DSS-induced colitis, which may be through alteration of BMDCs function.


Subject(s)
Colitis , Melatonin , MicroRNAs , RNA, Long Noncoding , Mice , Animals , Dextran Sulfate/toxicity , Dextran Sulfate/metabolism , Melatonin/pharmacology , Melatonin/therapeutic use , RNA-Seq , RNA, Long Noncoding/metabolism , RNA, Circular , Bone Marrow/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Colitis/chemically induced , Colitis/drug therapy , Colon/metabolism , Cytokines/metabolism , Disease Models, Animal , RNA, Messenger/metabolism , Dendritic Cells , MicroRNAs/genetics , MicroRNAs/metabolism , Mice, Inbred C57BL
8.
Cell Rep ; 40(12): 111361, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36130501

ABSTRACT

Hematopoietic stem/progenitor cells (HSPCs) are supported and regulated by niche cells in the bone marrow with an important characterization of physiological hypoxia. However, how hypoxia regulates HSPCs is still unclear. Here, we find that meteorin (Metrn) from hypoxic macrophages restrains HSPC mobilization. Hypoxia-induced factor 1α and Yin Yang 1 induce the high expression of Metrn in macrophages, and macrophage-specific Metrn knockout increases HSPC mobilization through modulating HSPC proliferation and migration. Mechanistically, Metrn interacts with its receptor 5-hydroxytryptamine receptor 2b (Htr2b) to regulate the reactive oxygen species levels in HSPCs through targeting phospholipase C signaling. The reactive oxygen species levels are reduced in HSPCs of macrophage-specific Metrn knockout mice with activated phospholipase C signaling. Targeting the Metrn/Htr2b axis could therefore be a potential strategy to improve HSPC mobilization for stem cell-based therapy.


Subject(s)
Bone Marrow Cells , Bone Marrow , Animals , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Hypoxia/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins , Reactive Oxygen Species/metabolism , Receptors, Serotonin/metabolism , Type C Phospholipases/metabolism
9.
Front Immunol ; 13: 846246, 2022.
Article in English | MEDLINE | ID: mdl-35983045

ABSTRACT

Inflammation has a role in the pathogenesis of childhood malnutrition. We investigated the effect of malnutrition and inflammatory challenge on bone marrow composition and bone health. We studied an established murine model of moderate acute malnutrition at baseline and after acute inflammatory challenge with bacterial lipopolysaccharide (LPS), a surrogate of Gram-negative bacterial sepsis, or Leishmania donovani, the cause of visceral leishmaniasis. Both of these infections cause significant morbidity and mortality in malnourished children. Of the 2 stimuli, LPS caused more pronounced bone marrow changes that were amplified in malnourished mice. LPS challenge led to increased inflammatory cytokine expression (Il1b, Il6, and Tnf), inflammasome activation, and inflammatory monocyte accumulation in the bone marrow of malnourished mice. Depletion of inflammatory monocytes in Csfr1-LysMcre-DT malnourished mice significantly reduced the inflammasome activation and IL1-ß production after LPS challenge. The inflammatory challenge also led to increased expansion of mesenchymal stem cells (MSCs), bone marrow adiposity, and expression of genes (Pparg, Adipoq, and Srbp1) associated with adipogenesis in malnourished mice. This suggests that inflammatory challenge promotes differentiation of BM MSCs toward the adipocyte lineage rather than toward bone-forming osteoblasts in the malnourished host. Concurrent with this reduced osteoblastic potential there was an increase in bone-resorbing osteoclasts, enhanced osteoclast activity, upregulation of inflammatory genes, and IL-1B involved in osteoclast differentiation and activation. The resulting weakened bone formation and increased bone resorption would contribute to the bone fragility associated with malnutrition. Lastly, we evaluated the effect of replacing lipid rich in omega-6 fatty acids (corn oil) with lipid-rich in omega-3 fatty acids (fish oil) in the nutrient-deficient diet. LPS-challenged malnourished mice that received dietary fish oil showed decreased expression of inflammatory cytokines and Rankl and reduced osteoclast differentiation and activation in the bone marrow. This work demonstrates that the negative effect of inflammatory challenge on bone marrow is amplified in the malnourished host. Increasing dietary intake of omega-3 fatty acids may be a means to reduce inflammation and improve bone health in malnourished children.


Subject(s)
Fatty Acids, Omega-3 , Malnutrition , Animals , Bone Density , Bone Marrow/metabolism , Cytokines/metabolism , Fish Oils , Inflammasomes , Inflammation , Lipopolysaccharides , Mice
10.
Biometals ; 35(4): 741-758, 2022 08.
Article in English | MEDLINE | ID: mdl-35635647

ABSTRACT

B-cell acute lymphoblastic leukemia (B-ALL) is a hematologic disorder characterized by the abnormal proliferation and accumulation of immature B-lymphoblasts arrested at various stages of differentiation. Despite advances in treatment, a significant percentage of pediatric patients with precursor B-ALL still relapse. Therefore, alternative therapies are needed to improve the cure rates for pediatric patients. TPEN (N, N, N', N'-tetrakis(2-pyridylmethyl)-ethylenediamine) is a pro-oxidant agent capable of selectively inducing apoptosis in leukemia cell lines. Consequently, it has been suggested that TPEN could be a potential agent for oxidative therapy. However, it is not yet known whether TPEN can selectively destroy leukemia cells in a more disease-like model, for example, the bloodstream and bone marrow (BM), ex vivo. This investigation is an extension of a previous study that dealt with the effect of TPEN on ex vivo isolated/purified refractory B-ALL cells. Here, we evaluated the effect of TPEN on whole BM from nonleukemic patients (control) or pediatric patients diagnosed with de novo B-ALL or refractory B-ALL cells by analyzing the hematopoietic cell lineage marker CD34/CD19. Although TPEN was innocuous to nonleukemic BM (n = 3), we found that TPEN significantly induced apoptosis in de novo (n = 5) and refractory B-ALL (n = 6) leukemic cell populations. Moreover, TPEN significantly increased the counts of cells positive for the oxidation of the stress sensor protein DJ-1, a sign of the formation of H2O2, and significantly increased the counts of cells positive for the pro-apoptotic proteins TP53, PUMA, and CASPASE-3 (CASP-3), indicative of apoptosis, in B-ALL cells. We demonstrate that TPEN selectively eliminates B-ALL cells (CD34 + /CD19 +) but no other cell populations in BM (CD34 + /CD19-; CD34-/CD19 + ; CD34-/CD19-) independent of age, diagnosis status (de novo or refractory), sex, karyotype, or immunophenotype. Understanding TPEN-induced cell death in leukemia cells provides insight into more effective therapeutic oxidation-inducing anticancer agents.


Subject(s)
Bone Marrow , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Antigens, CD19/metabolism , Bone Marrow/metabolism , Child , Ethylenediamines , Humans , Hydrogen Peroxide/metabolism , Immunophenotyping , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
11.
Nutrients ; 14(7)2022 Apr 06.
Article in English | MEDLINE | ID: mdl-35406143

ABSTRACT

Radiation therapy damages and depletes total bone marrow (BM) cellularity, compromising safety and limiting effective dosing. Aging also strains total BM and BM hematopoietic stem and progenitor cell (HSPC) renewal and function, resulting in multi-system defects. Interventions that preserve BM and BM HSPC homeostasis thus have potential clinical significance. Here, we report that 50% calorie restriction (CR) for 7-days or fasting for 3-days prior to irradiation improved mouse BM regrowth in the days and weeks post irradiation. Specifically, one week of 50% CR ameliorated loss of total BM cellularity post irradiation compared to ad libitum-fed controls. CR-mediated BM protection was abrogated by dietary sulfur amino acid (i.e., cysteine, methionine) supplementation or pharmacological inhibition of sulfur amino acid metabolizing and hydrogen sulfide (H2S) producing enzymes. Up to 2-fold increased proliferative capacity of ex vivo-irradiated BM isolated from food restricted mice relative to control mice indicates cell autonomy of the protective effect. Pretreatment with H2S in vitro was sufficient to preserve proliferative capacity by over 50% compared to non-treated cells in ex vivo-irradiated BM and BM HSPCs. The exogenous addition of H2S inhibited Ten eleven translocation 2 (TET2) activity in vitro, thus providing a potential mechanism of action. Short-term CR or fasting therefore offers BM radioprotection and promotes regrowth in part via altered sulfur amino acid metabolism and H2S generation, with translational implications for radiation treatment and aging.


Subject(s)
Hydrogen Sulfide , Radiation Injuries , Animals , Bone Marrow/metabolism , Caloric Restriction , Dietary Supplements , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Methionine/pharmacology , Mice , Mice, Inbred C57BL , Radiation, Ionizing
12.
J Biomed Mater Res A ; 110(4): 747-760, 2022 04.
Article in English | MEDLINE | ID: mdl-34713570

ABSTRACT

Recently, we have developed a hydroxyapatite (HAp)-hybridized double-network (DN) hydrogel (HAp/DN gel), which can robustly bond to the bone tissue in the living body. The purpose of this study is to clarify whether the HAp/DN gel surface can differentiate the bone marrow-derived mesenchymal stem cells (MSCs) to osteogenic cells. We used the MSCs which were harvested from the rabbit bone marrow and cultured on the polystyrene (PS) dish using the autogenous serum-supplemented medium. First, we confirmed the properties of MSCs by evaluating colony forming unit capacity, expression of MSC markers using flow cytometry, and multidifferential capacity. Secondly, polymerase chain reaction analysis demonstrated that the HAp/DN gel surface significantly enhanced mRNA expression of the eight osteogenic markers (TGF-ß1, BMP-2, Runx2, Col-1, ALP, OPN, BSP, and OCN) in the cultured MSCs at 7 days than the PS surfaces (p < 0.0001), while the DN gel and HAp surfaces provided no or only a slight effect on the expression of these markers except for Runx2. Additionally, the alkaline phosphatase activity was significantly higher in the cells cultured on the HAp/DN gel surface than in the other three material surfaces (p < 0.0001). Thirdly, when the HAp/DN gel plug was implanted into the rabbit bone marrow, MSC marker-positive cells were recruited in the tissue generated around the plug at 3 days, and Runx2 and OCN were highly expressed in these cells. In conclusion, this study demonstrated that the HAp/DN gel surface can differentiate the MSCs into osteogenic cells.


Subject(s)
Durapatite , Mesenchymal Stem Cells , Animals , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cells, Cultured , Durapatite/chemistry , Hydrogels/metabolism , Hydrogels/pharmacology , Osteogenesis/genetics , Rabbits
13.
Food Funct ; 12(24): 12800-12811, 2021 Dec 13.
Article in English | MEDLINE | ID: mdl-34859812

ABSTRACT

Dendritic cells (DCs) represent a heterogeneous family of immune cells that link innate and adaptive immunity and their activation is linked to metabolic changes that are essential to support their activity and function. Hence, targeting the metabolism of DCs represents an opportunity to modify the inflammatory and immune response. Among the natural matrices, Humulus lupulus (Hop) compounds have recently been shown to exhibit immunomodulatory and anti-inflammatory activity. This study aimed to evaluate the ability of specific Hop fractions to modulate DCs metabolism after stimulation with lipopolysaccharide (LPS) by an untargeted metabolomics approach and compare their effect with flavonol quercetin. Following liquid chromatography-based fractionation, three fractions (A, B, and C) were obtained and tested. Cytokine and gene expression were evaluated using ELISA and qPCR, respectively, while the untargeted metabolomics analysis was performed using a combined HILIC-HRMS and DI-FT-ICR approach. The HOP C fraction and quercetin could both reduce the production of several inflammatory cytokines such as IL-6, IL-1α, IL-1ß, and TNF, but differently from quercetin, the HOP C mechanism is independent of extracellular iron-sequestration and showed significant upregulation of the Nrf2/Nqo1 pathway and Ap-1 compared to quercetin. The untargeted analysis revealed the modulation of several key pathways linked to pro-inflammatory and glycolytic phenotypes. In particular, HOP C treatment could modulate the oxidative step of the pentose phosphate pathway (PPP) and reduce the inflammatory mediator succinate, citrulline, and purine-pyrimidine metabolism, differently from quercetin. These results highlight the potential anti-inflammatory mechanism of specific Hop-derived compounds in restoring the dysregulated metabolism in DCs, which can be used in preventive or adjuvant therapies to suppress the undesirable inflammatory response.


Subject(s)
Citrulline/metabolism , Dendritic Cells/metabolism , Humulus/metabolism , Inflammation/metabolism , Pyrimidines/metabolism , Quercetin/metabolism , Succinic Acid/metabolism , Animals , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/metabolism , Bone Marrow/immunology , Bone Marrow/metabolism , Citrulline/immunology , Dendritic Cells/immunology , Disease Models, Animal , Flavonoids , Humulus/immunology , Inflammation/immunology , Mass Spectrometry/methods , Metabolomics/methods , Mice , Mice, Inbred C57BL , Plant Extracts/immunology , Plant Extracts/metabolism , Purines , Pyrimidines/immunology , Quercetin/immunology , Succinic Acid/immunology
14.
Medicine (Baltimore) ; 100(43): e27646, 2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34713855

ABSTRACT

BACKGROUND: From the perspective of evidence-based medicine, the efficacy and safety of combined therapy for marrow suppression after chemotherapy is still unclear. Given that there is no high-quality meta-analysis to incorporate existing evidence, the purpose of this protocol is to design a systematically review and meta-analysis of the level I evidence to ascertain the efficacy and safety of acupuncture combined with traditional Chinese medicine preparation for marrow suppression after chemotherapy. METHODS: The following databases will be searched electronically by keyword combination mode: 4 British literature databases including PubMed, EMBASE, Scopus, and Cochrane Library, and 4 Chinese literature databases, including Chinese national knowledge infrastructure, VIP, and Wan fang database. The randomized controlled trials on acupuncture plus traditional Chinese medicine preparation for marrow suppression after chemotherapy will be included. The primary outcome is the elevation of hemoglobin, platelets, leukocytes, and neutrophils. The other outcomes include clinical symptoms, quality of life, and absolute value of reticulocyte. Risk bias analysis of the studies will be performed independently by 2 reviewers using the Cochrane Risk of Bias Assessment Tool. RESULTS: The review will add to the existing literature by showing compelling evidence and improved guidance in clinic settings. CONCLUSION: This protocol will provide a reliable theoretical basis for the following research.


Subject(s)
Acupuncture Therapy/methods , Antineoplastic Agents/adverse effects , Bone Marrow/metabolism , Medicine, Chinese Traditional/methods , Age Factors , Antineoplastic Agents/therapeutic use , Blood Platelets/metabolism , Combined Modality Therapy , Hemoglobins/metabolism , Humans , Leukocytes/metabolism , Neoplasm Staging , Neoplasms/drug therapy , Neutrophils/metabolism , Quality of Life , Randomized Controlled Trials as Topic , Research Design , Sex Factors , Meta-Analysis as Topic
15.
Diabetologia ; 64(11): 2575-2588, 2021 11.
Article in English | MEDLINE | ID: mdl-34430981

ABSTRACT

AIMS/HYPOTHESIS: Hypothalamic inflammation and sympathetic nervous system hyperactivity are hallmark features of the metabolic syndrome and type 2 diabetes. Hypothalamic inflammation may aggravate metabolic and immunological pathologies due to extensive sympathetic activation of peripheral tissues. Loss of somatostatinergic (SST) neurons may contribute to enhanced hypothalamic inflammation. METHODS: The present data show that leptin receptor-deficient (db/db) mice exhibit reduced hypothalamic SST neurons, particularly in the periventricular nucleus. We model this finding, using adeno-associated virus delivery of diphtheria toxin subunit A (DTA) driven by an SST-cre system to deplete these neurons in Sstcre/gfp mice (SST-DTA). RESULTS: SST-DTA mice exhibit enhanced hypothalamic c-Fos expression and brain inflammation as demonstrated by microglial and astrocytic activation. Bone marrow from SST-DTA mice undergoes skewed haematopoiesis, generating excess granulocyte-monocyte progenitors and increased proinflammatory (C-C chemokine receptor type 2; CCR2hi) monocytes. SST-DTA mice exhibited a 'diabetic retinopathy-like' phenotype: reduced visual function by optokinetic response (0.4 vs 0.25 cycles/degree; SST-DTA vs control mice); delayed electroretinogram oscillatory potentials; and increased percentages of retinal monocytes. Finally, mesenteric visceral adipose tissue from SST-DTA mice was resistant to catecholamine-induced lipolysis, displaying 50% reduction in isoprenaline (isoproterenol)-induced lipolysis compared with control littermates. Importantly, hyperglycaemia was not observed in SST-DTA mice. CONCLUSIONS/INTERPRETATION: The isolated reduction in hypothalamic SST neurons was able to recapitulate several hallmark features of type 2 diabetes in disease-relevant tissues.


Subject(s)
Adipose Tissue/metabolism , Bone Marrow/metabolism , Diabetes Mellitus, Type 2/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Retina/metabolism , Somatostatin/metabolism , Animals , Brain/metabolism , Diabetes Mellitus, Type 2/diagnosis , Diphtheria Toxin/toxicity , Electroretinography , Flow Cytometry , Immunohistochemistry , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
16.
Int J Mol Sci ; 22(13)2021 Jun 24.
Article in English | MEDLINE | ID: mdl-34202651

ABSTRACT

Dieting is a common but often ineffective long-term strategy for preventing weight gain. Similar to humans, adult rats exhibit progressive weight gain. The adipokine leptin regulates appetite and energy expenditure but hyperleptinemia is associated with leptin resistance. Here, we compared the effects of increasing leptin levels in the hypothalamus using gene therapy with conventional caloric restriction on weight gain, food consumption, serum leptin and adiponectin levels, white adipose tissue, marrow adipose tissue, and bone in nine-month-old female Sprague-Dawley rats. Rats (n = 16) were implanted with a cannula in the 3rd ventricle of the hypothalamus and injected with a recombinant adeno-associated virus, encoding the rat gene for leptin (rAAV-Lep), and maintained on standard rat chow for 18 weeks. A second group (n = 15) was calorically-restricted to match the weight of the rAAV-Lep group. Both approaches prevented weight gain, and no differences in bone were detected. However, calorically-restricted rats consumed 15% less food and had lower brown adipose tissue Ucp-1 mRNA expression than rAAV-Lep rats. Additionally, calorically-restricted rats had higher abdominal white adipose tissue mass, higher serum leptin and adiponectin levels, and higher marrow adiposity. Caloric restriction and hypothalamic leptin gene therapy, while equally effective in preventing weight gain, differ in their effects on energy intake, energy expenditure, adipokine levels, and body composition.


Subject(s)
Caloric Restriction , Energy Metabolism , Genetic Therapy , Hypothalamus/metabolism , Leptin/genetics , Adipokines/blood , Adipokines/genetics , Adipokines/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue, White/metabolism , Adiposity/genetics , Animals , Biomarkers , Body Weight , Bone Marrow/metabolism , Dependovirus/genetics , Energy Intake , Energy Metabolism/genetics , Female , Gene Expression , Genetic Therapy/methods , Genetic Vectors , Leptin/metabolism , Rats , Transgenes
17.
Int J Biol Sci ; 17(7): 1821-1836, 2021.
Article in English | MEDLINE | ID: mdl-33994865

ABSTRACT

Both osteoblasts and preosteoclasts contribute to the coupling of osteogenesis and angiogenesis, regulating bone regeneration. Astragaloside IV (AS-IV), a glycoside of cycloartane-type triterpene derived from the Chinese herb Astragalus membranaceus, exhibits various biological activities, including stimulating angiogenesis and attenuating ischemic-hypoxic injury. However, the effects and underlying mechanisms of AS-IV in osteogenesis, osteoclastogenesis, and bone regeneration remain poorly understood. In the present study, we found that AS-IV treatment inhibited osteoclastogenesis, preserved preosteoclasts, and enhanced platelet-derived growth factor-BB (PDGF-BB)-induced angiogenesis. Additionally, AS-IV promoted cell viability, osteogenic differentiation, and angiogenic gene expression in bone marrow mesenchymal stem cells (BMSCs). The activation of AKT/GSK-3ß/ß-catenin signaling was found to contribute to the effects of AS-IV on osteoclastogenesis and osteogenesis. Furthermore, AS-IV accelerated bone regeneration during distraction osteogenesis (DO), as evidenced from the improved radiological and histological manifestations and biomechanical parameters, accompanied by enhanced angiogenesis within the distraction zone. In summary, AS-IV accelerates bone regeneration during DO, by enhancing osteogenesis and preosteoclast-induced angiogenesis simultaneously, partially through AKT/GSK-3ß/ß-catenin signaling. These findings reveal that AS-IV may serve as a potential bioactive molecule for promoting the coupling of osteogenesis and angiogenesis, and imply that AKT/GSK-3ß/ß-catenin signaling may be a promising therapeutic target for patients during DO treatment.


Subject(s)
Bone Marrow/metabolism , Bone Regeneration/physiology , Neovascularization, Physiologic/physiology , Osteoblasts/drug effects , Osteogenesis/drug effects , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Bone Marrow/drug effects , Bone Regeneration/drug effects , Cell Proliferation , Cells, Cultured , Drugs, Chinese Herbal , Male , Models, Animal , Osteoblasts/cytology , Osteogenesis/physiology , Rats , Rats, Sprague-Dawley
18.
J Nutr ; 151(4): 800-809, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33693772

ABSTRACT

BACKGROUND: While cancer is common, its incidence varies widely by tissue. These differences are attributable to variable risk factors, such as environmental exposure, genetic inheritance, and lifetime number of stem cell divisions in a tissue. Folate deficiency is generally associated with increased risk for colorectal cancer (CRC) and acute lymphocytic leukemia (ALL). Conversely, high folic acid (FA) intake has also been associated with higher CRC risk. OBJECTIVE: Our objective was to compare the effect of folate intake on mutant frequency (MF) and types of mutations in the colon and bone marrow of mice. METHODS: Five-week-old MutaMouse male mice were fed a deficient (0 mg FA/kg), control (2 mg FA/kg), or supplemented (8 mg FA/kg) diet for 20 wk. Tissue MF was assessed using the lacZ mutant assay and comparisons made by 2-factor ANOVA. LacZ mutant plaques were sequenced using next-generation sequencing, and diet-specific mutation profiles within each tissue were compared by Fisher's exact test. RESULTS: In the colon, the MF was 1.5-fold and 1.3-fold higher in mice fed the supplemented diet compared with mice fed the control (P = 0.001) and deficient (P = 0.008) diets, respectively. This contrasted with the bone marrow MF in the same mice where the MF was 1.7-fold and 1.6-fold higher in mice fed the deficient diet compared with mice fed the control (P = 0.02) and supplemented (P = 0.03) diets, respectively. Mutation profiles and signatures (mutation context) were tissue-specific. CONCLUSIONS: Our data indicate that dietary folate intake affects mutagenesis in a tissue- and dose-specific manner in mice. Mutation profiles were generally tissue- but not dose-specific, suggesting that altered cellular folate status appears to interact with endogenous mutagenic mechanisms in each tissue to create a permissive context in which specific mutation types accumulate. These data illuminate potential mechanisms underpinning differences in observed associations between folate intake/status and cancer.


Subject(s)
Folic Acid/administration & dosage , Mutation Rate , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Colon/drug effects , Colon/metabolism , Dose-Response Relationship, Drug , Folic Acid/adverse effects , Folic Acid/blood , Folic Acid Deficiency/blood , Folic Acid Deficiency/genetics , High-Throughput Nucleotide Sequencing , Lac Operon/drug effects , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Mutagenesis , Organ Specificity
19.
J Bone Miner Metab ; 39(2): 163-173, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32889573

ABSTRACT

INTRODUCTION: The jabuticaba peel extract (JPE) contains bioactive compounds that regulate fat metabolism. Because the negative correlation between fat accumulation and bone formation in bone marrow, we hypothesized that JPE inhibits adipocyte as well as favors osteoblast differentiation of mesenchymal stromal cells (MSCs) under healthy and osteoporotic conditions, a disease that display an imbalance between adipocyte and osteoblast differentiation resulting in reduced bone mass. MATERIAL AND METHODS: To test these hypotheses, bone marrow MSCs were harvested from healthy and osteoporotic rats and cultured in adipogenic and osteogenic media with three concentrations of JPE, 0.25, 5 and 10 µg/ml, and vehicle (control). After selecting the most efficient concentrations of JPE, we used them to evaluate adipocyte and osteoblast differentiation of MSCs from both sources. RESULTS: We observed that, in general, JPE inhibited adipocyte differentiation of MSCs with more pronounced effects in cells from healthy than osteoporotic rats. In addition, JPE increased osteoblast differentiation, exhibiting a slightly higher osteogenic potential on MSCs from osteoporotic compared to healthy condition. CONCLUSION: Our results demonstrated that JPE drives MSCs to inhibit adipocyte differentiation and toward osteoblast differentiation under healthy and osteoporotic conditions. These findings pave the way for further translational studies to investigate the therapeutic possibilities of JPE in both prevention and treatment of osteoporosis.


Subject(s)
Adipocytes/cytology , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteoporosis/pathology , Plant Extracts/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Adipogenesis/physiology , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone and Bones/drug effects , Bone and Bones/pathology , Cell Differentiation/drug effects , Cells, Cultured , Female , Mesenchymal Stem Cells/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Osteoporosis/metabolism , Ovariectomy , Rats, Wistar
20.
Stem Cells Dev ; 30(1): 39-48, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33176587

ABSTRACT

A transgenic acute promyelocytic leukemia (APL) murine model established by Michael Bishop by cloning a human PML-RARα cDNA into the hMRP8 expression cassette has been widely used in the all-trans retinoid acid and arsenic preparations for the research of APL. However, in the existing literature, the data of regularity and characteristics of the pathogenesis of this model were still missing, which hinder the development of many studies, especially application of new technologies such as single-cell sequencing. Therefore, in this article, we have made up this part of the missing data using an improved APL murine model. We clarified the effects of different inoculation doses on the onset time, latency, morbidity, life span, and proportion of APL cells in peripheral blood (PB), spleen, bone marrow, and so on. The relationship between the proportion of APL cells in the bone marrow, spleen, and PB and organ histological changes was also revealed. These results were a supplement and refinement of this APL model. It would add to the knowledge base of the field and aid in ensuring that accurate models are used for directed interventions. It also provides a great convenience for the researchers who will carry out similar research.


Subject(s)
Disease Models, Animal , Leukemia, Promyelocytic, Acute/genetics , Oncogene Proteins, Fusion/genetics , Transgenes/genetics , Animals , Bone Marrow/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Flow Cytometry/methods , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Promyelocytic, Acute/blood , Leukemia, Promyelocytic, Acute/pathology , Male , Mice, Transgenic , Spleen/metabolism , Survival Analysis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL