Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Exp Mol Med ; 56(1): 118-128, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38200155

ABSTRACT

The harmful effects of fine particulate matter ≤2.5 µm in size (PM2.5) on human health have received considerable attention. However, while the impact of PM2.5 on the respiratory and cardiovascular systems has been well studied, less is known about the effects on stem cells in the bone marrow (BM). With an emphasis on the invasive characteristics of PM2.5, this review examines the current knowledge of the health effects of PM2.5 exposure on BM-residing stem cells. Recent studies have shown that PM2.5 enters the circulation and then travels to distant organs, including the BM, to induce oxidative stress, systemic inflammation and epigenetic changes, resulting in the reduction of BM-residing stem cell survival and function. Understanding the broader health effects of air pollution thus requires an understanding of the invasive characteristics of PM2.5 and its direct influence on stem cells in the BM. As noted in this review, further studies are needed to elucidate the underlying processes by which PM2.5 disturbs the BM microenvironment and inhibits stem cell functionality. Strategies to prevent or ameliorate the negative effects of PM2.5 exposure on BM-residing stem cells and to maintain the regenerative capacity of those cells must also be investigated. By focusing on the complex relationship between PM2.5 and BM-resident stem cells, this review highlights the importance of specific measures directed at safeguarding human health in the face of rising air pollution.


Subject(s)
Air Pollutants , Air Pollution , Mesenchymal Stem Cells , Humans , Particulate Matter/adverse effects , Air Pollutants/adverse effects , Bone Marrow , Air Pollution/adverse effects , Environmental Exposure
2.
Aging Dis ; 15(2): 755-766, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37548936

ABSTRACT

One of the most important strategies for successful aging is exercise. However, the effect of exercise can differ among individuals, even with exercise of the same type and intensity. Therefore, this study aims to confirm whether endurance training (ETR) has the same health-promoting effects on the musculoskeletal and hematopoietic systems regardless of age. Ten weeks of ETR improved endurance exercise capacity, with increased skeletal muscle mitochondrial enzymes in both young and old mice. In addition, age-related deterioration of muscle fiber size and bone microstructure was improved. The expression levels of myostatin, muscle RING-finger protein-1, and muscle atrophy F-box in skeletal muscle and peroxisome proliferator-activated receptor-γ in the femur increased with age but decreased after ETR. ETR differentially modulated hematopoietic stem cells (HSCs) depending on age; ETR induced HSC quiescence in young mice but caused HSC senescence in old mice. ETR has differential effects on modulation of the musculoskeletal and hematopoietic systems in old mice. In other words, endurance exercise is a double-edged sword for successful aging, and great effort is required to establish exercise strategies for healthy aging.


Subject(s)
Muscle, Skeletal , Transcription Factors , Mice , Animals , Transcription Factors/metabolism , Muscle, Skeletal/metabolism , Muscle Fibers, Skeletal/metabolism , Mitochondria/metabolism , Aging/physiology
3.
Aging Dis ; 14(3): 919-936, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37191410

ABSTRACT

Ionizing irradiation (IR) causes bone marrow (BM) injury, with senescence and impaired self-renewal of hematopoietic stem cells (HSCs), and inhibiting Wnt signaling could enhance hematopoietic regeneration and survival against IR stress. However, the underlying mechanisms by which a Wnt signaling blockade modulates IR-mediated damage of BM HSCs and mesenchymal stem cells (MSCs) are not yet completely understood. We investigated the effects of osteoblastic Wntless (Wls) depletion on total body irradiation (TBI, 5 Gy)-induced impairments in hematopoietic development, MSC function, and the BM microenvironment using conditional Wls knockout mutant mice (Col-Cre;Wlsfl/fl) and their littermate controls (Wlsfl/fl). Osteoblastic Wls ablation itself did not dysregulate BM frequency or hematopoietic development at a young age. Exposure to TBI at 4 weeks of age induced severe oxidative stress and senescence in the BM HSCs of Wlsfl/fl mice but not in those of the Col-Cre;Wlsfl/fl mice. TBI-exposed Wlsfl/fl mice exhibited greater impairments in hematopoietic development, colony formation, and long-term repopulation than TBI-exposed Col-Cre;Wlsfl/fl mice. Transplantation with BM HSCs or whole BM cells derived from the mutant, but not Wlsfl/fl mice, protected against HSC senescence and hematopoietic skewing toward myeloid cells and enhanced survival in recipients of lethal TBI (10 Gy). Unlike the Wlsfl/fl mice, the Col-Cre;Wlsfl/fl mice also showed radioprotection against TBI-mediated MSC senescence, bone mass loss, and delayed body growth. Our results indicate that osteoblastic Wls ablation renders BM-conserved stem cells resistant to TBI-mediated oxidative injuries. Overall, our findings show that inhibiting osteoblastic Wnt signaling promotes hematopoietic radioprotection and regeneration.

4.
J Hazard Mater ; 452: 131293, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37002998

ABSTRACT

Research on the negative impacts of PM2.5 have been focused on lung, brain, immune, and metabolism-related diseases. However, little is known about the mechanism underlying the effects of PM2.5 on the modulation of hematopoietic stem cell (HSC) fate. Maturation of the hematopoietic system and differentiation of hematopoietic stem progenitor cells (HSPCs) occurs soon after birth when infants are susceptible to external stresses. We investigated how exposure to atmospherically relevant artificial particulate matter of diameter < 2.5 µm (termed, PM2.5) affects HSPCs in newborns. The lungs of newborn mice exposed to PM2.5 exhibited higher levels of oxidative stress and inflammasome activation, which continued during aging. PM2.5 also stimulated oxidative stress and inflammasome activation in bone marrow (BM). PM2.5-exposed infant mice at 12 months but not at 6 months displayed progressive senescence of HSCs accompanied by preferential impairment of the BM microenvironment with age-related phenotypes, as evidenced by colony-forming assay and serial transplantation and animal survival experiments. Further, PM2.5-exposed middle-aged mice did not exhibit radioprotective potential. Collectively, exposure of newborns to PM2.5 causes progressive senescence of HSCs. These findings revealed a novel mechanism by which PM2.5 affects the fate of HSCs, highlighting the crucial role of early life exposure to air pollution in determining human health outcomes.


Subject(s)
Inflammasomes , Particulate Matter , Humans , Mice , Animals , Particulate Matter/toxicity , Hematopoietic Stem Cells , Oxidative Stress , Cell Differentiation
5.
Leukemia ; 37(4): 877-887, 2023 04.
Article in English | MEDLINE | ID: mdl-36707620

ABSTRACT

Studies of PrPC-derived prion disease generally focus on neurodegeneration. However, little is known regarding the modulation of hematopoietic stem progenitor cells (HSPCs) that express PrPC in prion infection. Among bone marrow (BM) hematopoietic cells, hematopoietic stem cells (HSCs) strongly express PrPC. A bioassay revealed the presence of misfolded prion protein (PrPSc) in BM cells derived from prion-infected mice; these BM cells demonstrated reproducible prion infectivity. At 5 months after infection with ME7, mice exhibited a significant decrease in the number of HSPCs. This decrease was mainly driven by increased apoptotic cell death, rather than cell cycle progression and senescence, in PrPC-positive but not PrPC-negative HSPC populations through a cell-autonomous mechanism. Notably, both PrPC-positive and PrPC-negative HSCs underwent cellular senescence, as indicated by high levels of senescence-associated factors and deficits in repopulation and self-renewal capacities at 7 months after infection. Senescence of HSCs occurred in the ME7-impaired BM microenvironment with aging phenotypes through non-cell autonomous mechanisms. These data provide novel evidence that prion infection differentially modulates HSC fate through both cell-autonomous and non-autonomous mechanisms.


Subject(s)
Prion Diseases , Prions , Mice , Animals , Prions/metabolism , Hematopoietic Stem Cells/metabolism , Prion Diseases/metabolism , Bone Marrow Cells/metabolism , Apoptosis
6.
Stem Cells ; 41(1): 93-104, 2023 01 30.
Article in English | MEDLINE | ID: mdl-36368017

ABSTRACT

While supplemental angiopoietin-1 (Ang1) improves hematopoiesis, excessive Ang1 induces bone marrow (BM) impairment, hematopoietic stem cell (HSC) senescence, and erythropoietic defect. Here, we examined how excessive Ang1 disturbs hematopoiesis and explored whether hematopoietic defects were related to its level using K14-Cre;c-Ang1 and Col2.3-Cre;c-Ang1 transgenic mice that systemically and locally overexpress cartilage oligomeric matrix protein-Ang1, respectively. We also investigated the impacts of Tie2 inhibitor and AMD3100 on hematopoietic development. Transgenic mice exhibited excessive angiogenic phenotypes, but K14-Cre;c-Ang1 mice showed more severe defects in growth and life span with higher presence of Ang1 compared with Col2.3-Cre;c-Ang1 mice. Dissimilar to K14-Cre;c-Ang1 mice, Col2.3-Cre;c-Ang1 mice did not show impaired BM retention or senescence of HSCs, erythropoietic defect, or disruption of the stromal cell-derived factor 1 (SDF-1)/CXCR4 axis. However, these mice exhibited a defect in platelet production depending on the expression of Tie2 and globin transcription factor 1 (GATA-1), but not GATA-2, in megakaryocyte progenitor (MP) cells. Treatment with Tie2 inhibitor recovered GATA-1 expression in MP cells and platelet production without changes in circulating RBC in transgenic mice. Consecutive AMD3100 administration not only induced irrecoverable senescence of HSCs but also suppressed formation of RBC, but not platelets, via correlated decreases in number of erythroblasts and their GATA-1 expression in B6 mice. Our results indicate that genetic overexpression of Ang1 impairs hematopoietic development depending on its level, in which megakaryopoiesis is preferentially impaired via activation of Ang1/Tie2 signaling, whereas erythropoietic defect is orchestrated by HSC senescence, inflammation, and disruption of the SDF-1/CXCR4 axis.


Subject(s)
Anemia , Thrombocytopenia , Mice , Animals , Cartilage Oligomeric Matrix Protein/genetics , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Mice, Transgenic , Anemia/genetics , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism
7.
Mater Sci Eng C Mater Biol Appl ; 135: 112673, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35581065

ABSTRACT

Scaffolds combined with bioactive agents can enhance bone regeneration at therapeutic sites. We explore whether combined supplementation with coumaric acid and recombinant human-cartilage oligomeric matrix protein-angiopoietin 1 (rhCOMP-Ang1) is an ideal approach for bone tissue engineering. We developed coumaric acid-conjugated absorbable collagen scaffold (CA-ACS) and investigated whether implanting CA-ACS in combination with rhCOMP-Ang1 facilitates ACS- or CA-ACS-mediated bone formation using a rat model of critically sized mandible defects. We examined the mechanisms by which coumaric acid and rhCOMP-Ang1 regulate behaviors of human periodontal ligament fibroblasts (hPLFs). The CA-ACS exhibits greater anti-degradation and mechanical strength properties than does ACS alone. Implanting CA-ACS loaded with rhCOMP-Ang1 greatly enhances bone regeneration at the defect via the activation of angiogenic, osteogenic, and anti-osteoclastic responses compared with other rat groups implanted with an ACS alone or CA-ACS. Treatment with both rhCOMP-Ang1 and coumaric acid increases proliferation, mineralization, and migration of cultured hPLFs via activation of the Ang1/Tie2 signaling axis at a greater rate than treatment with either of them alone. Collectively, this study demonstrates that CA-ACS impregnated with rhCOMP-Ang1 enhances bone regeneration at therapeutic sites, and this enhancement is associated with a synergistic interaction between rhCOMP-Ang1-mediated angiogenesis and coumaric acid-related antioxidant responses.


Subject(s)
Angiopoietin-1 , Antioxidants , Angiopoietin-1/metabolism , Angiopoietin-1/pharmacology , Animals , Antioxidants/pharmacology , Cartilage Oligomeric Matrix Protein , Collagen/pharmacology , Coumaric Acids , Mandible , Rats
8.
Antioxidants (Basel) ; 10(8)2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34439457

ABSTRACT

While total body irradiation (TBI) is an everlasting curative therapy, the irradiation can cause long-term bone marrow (BM) injuries, along with senescence of hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) via reactive oxygen species (ROS)-induced oxidative damages. Thus, ameliorating or preventing ROS accumulation and oxidative stress is necessary for TBI-requiring clinical treatments. Here, we explored whether administration of ferulic acid, a dietary antioxidant, protects against TBI-mediated systemic damages, and examined the possible mechanisms therein. Sublethal TBI (5 Gy) decreased body growth, lifespan, and production of circulating blood cells in mice, together with ROS accumulation, and senescence induction of BM-conserved HSCs and MSCs. TBI also impaired BM microenvironment and bone mass accrual, which was accompanied by downregulated osteogenesis and by osteoclastogenic and adipogenic activation in BM. Long-term intraperitoneal injection of ferulic acid (50 mg/kg body weight, once per day for 37 consecutive days) protected mice from TBI-mediated mortality, stem cell senescence, and bone mass loss by restoring TBI-stimulated disorders in osteogenic, osteoclastic, and adipogenic activation in BM. In vitro experiments using BM stromal cells supported radioprotective effects of ferulic acid on TBI-mediated defects in proliferation and osteogenic differentiation. Overall, treatment with ferulic acid prevented TBI-mediated liver damage and enhanced endogenous antioxidant defense systems in the liver and BM. Collectively, these results support an efficient protection of TBI-mediated systemic defects by supplemental ferulic acid, indicating its clinical usefulness for TBI-required patients.

9.
Mol Cells ; 44(4): 254-266, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33935045

ABSTRACT

Numerous studies highlight the potential benefits potentials of supplemental cartilage oligomeric matrix protein-angiopoietin-1 (COMP-Ang1) through improved angiogenic effects. However, our recent findings show that excessive overexpression of COMP-Ang1 induces an impaired bone marrow (BM) microenvironment and senescence of hematopoietic stem cells (HSCs). Here, we investigated the underlying mechanisms of how excessive COMP-Ang1 affects the function of BM-conserved stem cells and hematopoiesis using K14-Cre;inducible-COMP-Ang1-transgenic mice. Excessive COMP-Ang1 induced peripheral egression and senescence of BM HSCs and mesenchymal stem cells (MSCs). Excessive COMP-Ang1 also caused abnormal hematopoiesis along with skewed differentiation of HSCs toward myeloid lineage rather than lymphoid lineage. Especially, excessive COMP-Ang1 disturbed late-stage erythroblast maturation, followed by decreased expression of stromal cell-derived factor 1 (SDF-1) and globin transcription factor 1 (GATA-1) and increased levels of superoxide anion and p-p38 kinase. However, transplantation with the mutant-derived BM cells or treatment with rhCOMP-Ang1 protein did not alter the frequency or GATA-1 expression of erythroblasts in recipient mice or in cultured BM cells. Together, our findings suggest that excessive COMP-Ang1 impairs the functions of BM HSCs and MSCs and hematopoietic processes, eventually leading to abnormal erythropoiesis via imbalanced SDF-1/CXCR4 axis and GATA-1 expression rather than Ang1/Tie2 signaling axis alterations.


Subject(s)
Angiopoietin-1/metabolism , Erythrocytes/metabolism , Hematopoiesis/genetics , Animals , Cell Differentiation , Humans , Mice , Mice, Transgenic
10.
Stem Cells ; 39(1): 103-114, 2021 01.
Article in English | MEDLINE | ID: mdl-33038284

ABSTRACT

Although functional association between Wnt signaling and bone homeostasis has been well described through genetic ablation of Wntless (Wls), the mechanisms of how osteoblastic Wls regulates the fate of bone marrow stromal cells (BMSCs) and hematopoietic stem cells (HSCs) in relation to age are not yet understood. Here, we generated Col2.3-Cre;Wlsfl/fl mice that were free from premature lethality and investigated age-related impacts of osteoblastic Wls deficiency on hematopoiesis, BM microenvironment, and maintenance of BMSCs (also known as BM-derived mesenchymal stem/stromal cells) and HSCs. Ablation of osteoblastic Wls deteriorated BM microenvironment and bone mass accrual along with age-independent effects on functions of BMSCs. Osteoblastic Wls deletion impaired HSC repopulation and progeny with skewing toward myeloid lineage cells only at old stage. As proven by hallmarks of stem cell senescence, osteoblastic Wls ablation differentially induced senescence of BMSCs and HSCs in relation to age without alteration in their BM frequency. Our findings support that deletion of Wls in Col2.3-expressing cells induces senescence of BMSCs and impairs BM microenvironment in age-independent manner. Overall, long-term deterioration in BM microenvironment contributes to age-related HSC senescence with impaired progeny and hematopoiesis, which also suggests possible roles of osteoblastic Wls on the maintenance of BM HSCs.


Subject(s)
Aging/metabolism , Bone Marrow Cells/metabolism , Gene Deletion , Osteoblasts/metabolism , Receptors, G-Protein-Coupled/deficiency , Stem Cells/metabolism , Animals , Mice , Mice, Transgenic , Receptors, G-Protein-Coupled/metabolism
12.
Aging Dis ; 10(6): 1320-1327, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31788343

ABSTRACT

Total body irradiation (TBI) serves as an effectively curative therapy for cancer patients and adversely causes long-term residual bone marrow (BM) injury with premature senescence of hematopoietic stem cells (HSCs), which is mediated by increased production of reactive oxygen species (ROS). In the present study, we investigated how the exposure time of TBI in a mouse model affects HSCs and whether the treatment of caffeic acid (CA), a known dietary phenolic antioxidant, has a radioprotective effect. Single (S)-TBI at a sublethal dose (5 Gy) caused relatively higher induction of mitochondrial ROS and senescence-related factors in HSCs than those in hematopoietic progenitor cells (HPCs) and Lineage-Sca-1+c-Kit+ (LSK) cells, as well as reduced clonogenic formation and donor cell-derived reconstituting capacity. Repetitive double (D)-TBI (two months after the S-TBI at a dose of 5Gy) further weakened HSPC function via mitochondrial ROS accumulation and senescence-associated ß-galactosidase (SA-ß-gal) activity. Oral administration of CA (20 mg/kg) five times before and once immediately after TBI ameliorated ROS generation and TBI-induced HSC senescence and its radioprotective effect was long lasting in S-TBI mice but not in D-TBI mice. Further, supplementation of CA also induced apoptotic cell death of colon cancer cells. Collectively, these findings indicate that CA has a dual effect, ameliorating HSC senescence-accompanied long-term BM injury in S-TBI mice and stimulating apoptotic cell death of colon cancer cells.

13.
Biochem Biophys Res Commun ; 499(3): 669-674, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29604278

ABSTRACT

Supplemental Angiopoietin 1 (Ang1) exerts its therapeutic potential on microvascular regression-associated diseases, and this potential is linked with the function of hematopoietic stem cells (HSCs). However, the underlying mechanisms of the effect of enhanced angiogenesis on the modulation of HSCs are not yet defined. Here, we generated transgenic mice expressing Cartilage Oligomeric Matrix Protein (COMP)-Ang1 in keratin 14-expressing cells. The mutant animals expressed excessive angiogenic characteristics in the skin and bone marrow (BM) along with redder skin with more numerous and branched vessels compared with their wild-type (WT) littermates. The mutants displayed reduced long bone formation and osteoclast activity than did WT littermates and had fewer CD150+CD48-Lineage-Sca-1+c-Kit+ (LSK) cells in the BM. The mutants also exhibited greater senescence-associated (SA) ß-gal activity, p16INK4a protein expression, and superoxide anion levels in CD150+CD48-LSK cells in the BM. Furthermore, transplantation assay revealed that the mutant-derived LSK cells were inferior to the cells derived from WT littermate in inducing competitive repopulating capacity in the recipients. Collectively, our results demonstrate that persistent and prolonged administration of COMP-Ang1 by inducible transgenic expression mediates excessive angiogenesis in the body and impairs BM microenvironment, eventually leading to senescence of BM HSCs.


Subject(s)
Angiopoietin-1/genetics , Bone Marrow/metabolism , Cartilage Oligomeric Matrix Protein/genetics , Cellular Microenvironment , Cellular Senescence , Gene Expression , Hematopoietic Stem Cells/metabolism , Recombinant Fusion Proteins/genetics , Animals , Cartilage Oligomeric Matrix Protein/metabolism , Hematopoietic Stem Cells/cytology , Humans , Mice, Transgenic , Mutation/genetics , Neovascularization, Physiologic , Osteoclasts , Recombinant Fusion Proteins/metabolism
14.
Mol Cell Biochem ; 419(1-2): 157-63, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27431005

ABSTRACT

Nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/heme oxygenase-1 (HO-1) signal is known to play important roles in controlling bone homeostasis. This study examined how oxidative stress affects the mineralization of embryonic stem (ES) cells by exposing them to glucose oxidase (GO), which continuously generates H2O2 at low concentrations. The roles of Nrf2/HO-1 and mitogen-activated protein kinases on osteogenesis in GO-exposed ES cells were also investigated. GO treatment at relatively low concentrations did not change the viability of ES cells, whereas it enhanced osteogenic differentiation and mineralization in the cells. GO treatment (1 mU/ml) augmented the induction of runt-related transcription factor 2 (Runx2), Nrf2, and HO-1 in ES cells. GO-mediated acceleration of Runx2 expression and mineralization was inhibited either by Nrf2 knockdown or by treating with 5 µM PD98059, an inhibitor of phospho-extracellular signal-regulated kinase (p-ERK). The GO-stimulated mineralization was also suppressed by treating the cells with reduced glutathione or catalase, but not by superoxide dismutase or N-acetyl-cysteine. Collectively, our results demonstrate that a mild oxidative stress activates Nrf2/HO-1 signaling and an ERK-mediated pathway, and facilitates the mineralization of ES cells with a corresponding increase in Runx2.


Subject(s)
Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Glucose Oxidase/pharmacology , MAP Kinase Signaling System/drug effects , Mouse Embryonic Stem Cells/metabolism , NF-E2-Related Factor 2/metabolism , Osteogenesis/drug effects , Animals , Cell Line , Core Binding Factor Alpha 1 Subunit/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Oxidative Stress/drug effects
15.
Stem Cells ; 34(10): 2601-2607, 2016 10.
Article in English | MEDLINE | ID: mdl-27300755

ABSTRACT

Osteoclasts form a bone marrow (BM) cavity serving as a hematopoietic niche for the maintenance of hematopoietic stem cells (HSCs). However, the role of osteoclasts in the BM has been controversially reported and remains to be further understood. In the present study, we investigated how osteoclasts affect the modulation of hematopoietic stem/progenitor cells in the BM by administering bisphosphate alendronate (ALN) to B6 mice for 21 consecutive days to inhibit osteoclast activity. ALN treatment caused a reduction in the number of tartrate-resistant acid phosphate (TRAP)-positive osteoclast cells and an increase in bone mineral density, particularly in the trabecular zone, but not in the cortical zone of the BM. Osteoclast inhibition caused by ALN treatment decreased mitochondrial reactive oxygen species (ROS) generation and SA-ß-gal activity of CD150+ CD48- Lineage-Sca-1+ c-Kit+ (LSK) cells, eventually leading to an improvement in the engraftment potential and self-renewal activity of HSCs. Moreover, ALN-treated mice exhibited an enhanced resistance of HSCs in response to the genotoxic stress of 5-fluorouracil, as determined by mitochondrial ROS generation, SA-ß-gal activity, and p16INK4a expression in subsets of LSK and CD150+ CD48- LSK cells as well as competitive assay. Collectively, our findings indicate that inhibition of osteoclast activity improves the long-term engraftment potential and stress resistance of HSCs. Stem Cells 2016;34:2601-2607.


Subject(s)
Alendronate/administration & dosage , Alendronate/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Osteoclasts/metabolism , Stress, Physiological , Animals , Antineoplastic Agents/adverse effects , Bone Density/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cancellous Bone/drug effects , Cancellous Bone/physiology , Cell Self Renewal/drug effects , Female , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Mice, Inbred C57BL , Osteoclasts/drug effects , Peripheral Blood Stem Cells/cytology , Spleen/cytology , Stress, Physiological/drug effects , Time Factors
16.
Mater Sci Eng C Mater Biol Appl ; 44: 52-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25280679

ABSTRACT

Tissue engineering requires functional polymeric membrane for adequate space for cell migration and attachment within the nanostructure. Therefore, biocompatible propolis loaded polyurethane (propolis/PU) nanofibers were successfully prepared using electrospinning of propolis/PU blend solution. Here, composite nanofibers were subjected to detailed analysis using electron microscopy, FT-IR spectroscopy, thermal gravimetric analysis (TGA), and mechanical properties and water contact angle measurement. FE-SEM images revealed that the composite nanofibers became point-bonded with increasing amounts of propolis in the blend due to its adhesive properties. Incorporation of small amount of propolis through PU matrix could improve the hydrophilicity and mechanical strength of the fibrous membrane. In order to assay the cytocompatibility and cell behavior on the composite scaffolds, fibroblast cells were seeded on the matrix. Results suggest that the incorporation of propolis into PU fibers could increase its cell compatibility. Moreover, composite nanofibers have effective antibacterial activity. Therefore, as-synthesized nanocomposite fibrous mat has great potentiality in wound dressing and skin tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Nanofibers/chemistry , Polyurethanes/chemistry , Propolis/chemistry , 3T3-L1 Cells , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biocompatible Materials/pharmacology , Cell Survival/drug effects , Chemical Phenomena , Escherichia coli/drug effects , Escherichia coli/growth & development , Fibroblasts/chemistry , Hydrophobic and Hydrophilic Interactions , Mechanical Phenomena , Mice , Microscopy, Electron , Nanocomposites/chemistry , Polymers , Polyurethanes/pharmacology , Propolis/pharmacology , Spectroscopy, Fourier Transform Infrared , Tissue Engineering
SELECTION OF CITATIONS
SEARCH DETAIL
...