Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 376
Filter
Add more filters

Publication year range
1.
Mol Biol Rep ; 51(1): 596, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38683461

ABSTRACT

BACKGROUND: Arnica montana and Bellis perennis are two medicinal plants that are thought to accelerate bone repair in homoeopathic literature. Mesenchymal stem cells (MSCs) are multipotent stem cells with the ability to differentiate and regenerate bone or osteogenesis. Hence, we aimed to determine the role of Arnica montana and Bellis perennis on the osteogenic differentiation of the C3H10T1/2 stem cell line. METHODS AND RESULTS: The cell proliferation of Arnica montana and Bellis perennis was evaluated by MTT assay. Osteogenic differentiation of C3H10T1/2 was induced by the addition of ß-glycerophosphate, ascorbic acid and dexamethasone in the differentiation medium over 3 weeks. Cells were treated with Arnica montana and Bellis perennis individually as well as in combination. The osteogenic differentiation potential of Arnica montana and Bellis perennis to differentiate C3H10T1/2 into osteoblasts was measured by alkaline phosphatase activity, alizarin red staining and the expression of Osteocalcin using immunostaining and qRT-PCR. Arnica montana and Bellis perennis could enhance C3H10T1/2 cell proliferation at 1600 µg. Further, the compound showed the ability to augment osteogenesis as confirmed by increased expression of alkaline phosphatase and enhanced calcium accumulation as seen by the Alizarin Red staining and quantification. Enhanced osteogenesis was further supported by the increased expression of osteocalcin in the treated cells with individual and combined doses of Arnica montana and Bellis perennis. Therefore, the findings provide additional support for the positive impact of Arnica montana and Bellis perennis on bone formation. CONCLUSIONS: Our findings suggest that homoeopathic compounds Arnica montana and Bellis perennis can augment osteogenesis individually as well as in combination.


Subject(s)
Arnica , Cell Differentiation , Cell Proliferation , Mesenchymal Stem Cells , Osteogenesis , Plant Extracts , Osteogenesis/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Cell Differentiation/drug effects , Animals , Cell Proliferation/drug effects , Mice , Plant Extracts/pharmacology , Cell Line , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/cytology , Alkaline Phosphatase/metabolism , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Osteocalcin/metabolism , Osteocalcin/genetics
2.
Br J Haematol ; 193(2): 410-414, 2021 04.
Article in English | MEDLINE | ID: mdl-33216370

ABSTRACT

Eltrombopag has been added to first-line treatment of immune aplastic anaemia (AA), resulting in higher responses. We analysed marrow samples of AA patients who responded to immunosuppressive therapy (IST) alone or in combination with eltrombopag for the composition of the haematopoietic stem and progenitor cell (HSPC) compartment. The number of CD34+ cells and multipotent progenitors was higher in patients treated with eltrombopag (P < 0·005; P < 0·05; respectively), but not the number of stem cells. No aberrant phenotype was observed. These results indicate that eltrombopag augments CD34+ cells in vivo and preferentially expands multipotent progenitors, but not stem cells.


Subject(s)
Anemia, Aplastic/drug therapy , Benzoates/pharmacology , Hematopoietic Stem Cells/drug effects , Hydrazines/pharmacology , Multipotent Stem Cells/drug effects , Pyrazoles/pharmacology , Receptors, Thrombopoietin/agonists , Adolescent , Adult , Antigens, CD34/drug effects , Benzoates/administration & dosage , Biopsy, Needle/methods , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Brazil/epidemiology , Female , Flow Cytometry/methods , Hematopoietic Stem Cells/cytology , Humans , Hydrazines/administration & dosage , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Multipotent Stem Cells/cytology , Pyrazoles/administration & dosage
3.
Stem Cells ; 38(9): 1159-1174, 2020 09.
Article in English | MEDLINE | ID: mdl-32442338

ABSTRACT

C-X-C motif chemokine ligand 12 (CXCL12; aka SDF1α) is a major regulator of a number of cellular systems, including hematopoiesis, where it influences hematopoietic cell trafficking, proliferation, and survival during homeostasis and upon stress and disease. A variety of constitutive, temporal, ubiquitous, and cell-specific loss-of-function models have documented the functional consequences on hematopoiesis upon deletion of Cxcl12. Here, in contrast to loss-of-function experiments, we implemented a gain-of-function approach by generating a doxycycline-inducible transgenic mouse model that enables spatial and temporal overexpression of Cxcl12. We demonstrated that ubiquitous CXCL12 overexpression led to an increase in multipotent progenitors in the bone marrow and spleen. The CXCL12+ mice displayed reduced reconstitution potential as either donors or recipients in transplantation experiments. Additionally, we discovered that Cxcl12 overexpression improved hematopoietic stem and progenitor cell mobilization into the blood, and conferred radioprotection by promoting quiescence. Thus, this new CXCL12+ mouse model provided new insights into major facets of hematopoiesis and serves as a versatile resource for studying CXCL12 function in a variety of contexts.


Subject(s)
Chemokine CXCL12/metabolism , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Radiation Protection , Animals , Benzylamines/pharmacology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Count , Cell Cycle/drug effects , Cyclams/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Neovascularization, Physiologic/drug effects
4.
Purinergic Signal ; 17(4): 681-691, 2021 12.
Article in English | MEDLINE | ID: mdl-34351588

ABSTRACT

The ability of cardiac adipose-derived stem cells (cADSC) to differentiate into multiple cell types has opened new perspectives in cardiac cell-based regenerative therapies. P2Y nucleotide receptors have already been described as regulators of adipogenic differentiation of cADSC and bone marrow-derived stem cells. In this study, we defined UTP as a regulator of cADSC endothelial differentiation. A daily UTP stimulation of cADSC during endothelial predifferentiation increased their capacity to form an endothelial network in matrigel. Additionally, pro-angiogenic UTP target genes such as epiregulin and hyaluronan synthase-1 were identified in predifferentiated cADSC by RNA sequencing experiments. Their regulation by UTP was confirmed by qPCR and ELISA experiments. We then evaluated the capacity of UTP-treated predifferentiated cADSC to increase post-ischemic revascularization in mice subjected to left anterior descending artery ligation. Predifferentiated cADSC treated or not with UTP were injected in the periphery of the infarcted zone, 3 days after ligation. We observed a significant increase of capillary density 14 and 30 days after UTP-treated predifferentiated cADSC injection, correlated with a reduction of cardiac fibrosis. This revascularization increase was not observed after injection of UTP-treated cADSC deficient for UTP and ATP nucleotide receptor P2Y2. The present study highlights the P2Y2 receptor as a regulator of cADSC endothelial differentiation and as a potential target for the therapeutic use of cADSC in post-ischemic heart revascularization.


Subject(s)
Cell Differentiation/drug effects , Multipotent Stem Cells/drug effects , Neovascularization, Physiologic/drug effects , Uridine Triphosphate/pharmacology , Animals , Epiregulin/genetics , Epiregulin/metabolism , Mice , Mice, Knockout , Multipotent Stem Cells/metabolism , Receptors, Purinergic P2Y2/genetics , Receptors, Purinergic P2Y2/metabolism
5.
Differentiation ; 112: 67-76, 2020.
Article in English | MEDLINE | ID: mdl-32045848

ABSTRACT

To induce and maintain naïve pluripotency in mouse embryonic and induced pluripotent stem cells (ESCs/iPSCs), chemically defined N2B27 medium with PD0325901, CHIR99021, and leukemia inhibitory factor (2i/LIF) is a classic and simple condition. However, this method cannot be simply extrapolated to human ESCs/iPSCs that are principally stabilized in primed pluripotency and become primitive neuroepithelium-like cells in N2B27+2i/LIF culture. Here, we assessed iPSC reprogramming of fibroblasts from chimpanzee, our closest living relative, in N2B27+2i/LIF culture. Under this condition, chimpanzee cells formed alkaline phosphatase-positive dome-shaped colonies. The colony-forming cells could be stably expanded by serial passaging without a ROCK inhibitor. However, their gene expression was distinct from iPSCs and neuroepithelium. They expressed the OCT3/4 transgene and a subset of transcripts associated with pluripotency, mesenchymal-epithelial transition, and neural crest formation. These cells exhibited a differentiation potential into the three germ layers in vivo and in vitro. The current study demonstrated that iPSC reprogramming in N2B27+2i/LIF culture converted chimpanzee fibroblasts into a multipotent cancerous state with unique gene expression, but not fully pluripotent stem cells.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Multipotent Stem Cells/cytology , Animals , Benzamides/pharmacology , Cell Differentiation/drug effects , Cellular Reprogramming/drug effects , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Epithelial-Mesenchymal Transition/genetics , Fibroblasts/cytology , Fibroblasts/drug effects , Germ Layers/drug effects , Germ Layers/growth & development , Humans , Leukemia Inhibitory Factor/pharmacology , Mice , Multipotent Stem Cells/drug effects , Neural Crest/cytology , Pan troglodytes , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology
6.
Molecules ; 26(6)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33809391

ABSTRACT

Stem cells from apical papilla (SCAPs) are desirable sources of dentin regeneration. Epigallocatechin-3-gallate (EGCG), a natural component of green tea, shows potential in promoting the osteogenic differentiation of bone mesenchymal stem cells. However, whether EGCG regulates the odontogenic differentiation of SCAPs and how this occurs remain unknown. SCAPs from immature human third molars (16-20 years, n = 5) were treated with a medium containing different concentrations of EGCG or bone morphogenic protein 2 (BMP2), with or without LDN193189 (an inhibitor of the canonical BMP pathway). Cell proliferation and migration were analyzed using a CCK-8 assay and wound-healing assay, respectively. Osteo-/odontogenic differentiation was evaluated via alkaline phosphatase staining, alizarin red S staining, and the expression of osteo-/odontogenic markers using qPCR and Western blotting. We found that EGCG (1 or 10 µM) promoted the proliferation of SCAPs, increased alkaline phosphatase activity and mineral deposition, and upregulated the expression of osteo-/odontogenic markers including dentin sialophosphoprotein (Dspp), dentin matrix protein-1 (Dmp-1), bone sialoprotein (Bsp), and Type I collagen (Col1), along with the elevated expression of BMP2 and phosphorylation level of Smad1/5/9 (p < 0.01). EGCG at concentrations below 10 µM had no significant influence on cell migration. Moreover, EGCG-induced osteo-/odontogenic differentiation was significantly attenuated via LDN193189 treatment (p < 0.01). Furthermore, EGCG showed the ability to promote mineralization comparable with that of recombinant BMP2. Our study demonstrated that EGCG promotes the osteo-/odontogenic differentiation of SCAPs through the BMP-Smad signaling pathway.


Subject(s)
Catechin/analogs & derivatives , Dental Papilla/cytology , Dental Papilla/drug effects , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Odontogenesis/drug effects , Osteogenesis/drug effects , Adolescent , Biomarkers/metabolism , Bone Morphogenetic Protein 2/metabolism , Catechin/pharmacology , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dental Papilla/metabolism , Humans , Multipotent Stem Cells/metabolism , Regeneration/drug effects , Signal Transduction/drug effects , Smad Proteins/metabolism , Young Adult
7.
Bull Exp Biol Med ; 172(2): 175-179, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34853967

ABSTRACT

In 3-month bone marrow transplants of CBA mice from bone marrow donors receiving single injections of TLR-4 ligand (LPS) or NOD-2 ligand (muramyl dipeptide, MDP) 24 h before transplantation, an increase in the total number of MSCs (by 2.6 and 1.9 times, respectively), as well as a slight increase in the number of nuclear cells and the mass of bone capsules (by 1.3 and 1.2 times) were observed. After combined administration of MDР and LPS to donors, the total content of MSCs in the grafts was higher by 1.6 times in comparison with the total result of their isolated administration (and by 7.2 times in comparison with the control). At the same time, the concentration of osteogenic MSCs in the grafts of all groups was almost the same and corresponded to the control level. The number of nuclear cells and the mass of bone capsules of the grafts after combined administration of LPS and MDP were close (~80%) to the sum of the results of their isolated administration. These findings suggest that activation of the stromal tissue and the success of bone marrow transplantation depend on the intensity of innate immune responses. These data can be useful for the development of optimal methods of tissue transplantation.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/administration & dosage , Bone Marrow Cells/drug effects , Bone Marrow Transplantation , Lipopolysaccharides/administration & dosage , Tissue Donors , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Count , Cell Proliferation/drug effects , Cells, Cultured , Drug Combinations , Lipopolysaccharides/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred CBA , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Nod2 Signaling Adaptor Protein/agonists , Toll-Like Receptor 4/agonists
8.
Bull Exp Biol Med ; 171(3): 333-337, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34297290

ABSTRACT

We studied the participation of JNK and p53 in the realization of the growth potential of different types of progenitors of the subventricular zone of mouse brain and secretion of neurotrophins by glial cells. The stimulating role of these signaling molecules in mitotic activity and specialization of multipotent neural stem cells was shown. It was found that JNK and p53 do not participate in the regulation of committed neuronal progenitor cells (clonogenic PSA-NCAM+ cells). A dependence of neurotrophic growth factors in individual populations of neuroglia on activity of these protein kinase and transcription factor was revealed. The role of JNK and p53 in astrocytes consists in stimulation of their secretion, and in microglial cells, on the contrary, in its inhibition. The secretory neurotrophic function of oligodendrogliocytes is not associated with JNK and p53 activity.


Subject(s)
Astrocytes/metabolism , MAP Kinase Kinase 4/genetics , Multipotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Neuroglia/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Astrocytes/cytology , Astrocytes/drug effects , Benzothiazoles/pharmacology , CD56 Antigen/genetics , CD56 Antigen/metabolism , Culture Media, Conditioned/pharmacology , Gene Expression Regulation , Lateral Ventricles/cytology , Lateral Ventricles/drug effects , Lateral Ventricles/metabolism , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neuroglia/cytology , Neuroglia/drug effects , Signal Transduction , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/metabolism
9.
J Cell Mol Med ; 24(1): 886-898, 2020 01.
Article in English | MEDLINE | ID: mdl-31742908

ABSTRACT

MG53 is an important membrane repair protein and partially protects bone marrow multipotent adult progenitor cells (MAPCs) against oxidized low-density lipoprotein (ox-LDL). The present study was to test the hypothesis that the limited protective effect of MG53 on MAPCs was due to ox-LDL-induced reduction of MG53. MAPCs were cultured with and without ox-LDL (0-20 µg/mL) for up to 48 hours with or without MG53 and antioxidant N-acetylcysteine (NAC). Serum MG53 level was measured in ox-LDL-treated mice with or without NAC treatment. Ox-LDL induced significant membrane damage and substantially impaired MAPC survival with selective inhibition of Akt phosphorylation. NAC treatment effectively prevented ox-LDL-induced reduction of Akt phosphorylation without protecting MAPCs against ox-LDL. While having no effect on Akt phosphorylation, MG53 significantly decreased ox-LDL-induced membrane damage and partially improved the survival, proliferation and apoptosis of MAPCs in vitro. Ox-LDL significantly decreased MG53 level in vitro and serum MG53 level in vivo without changing MG53 clearance. NAC treatment prevented ox-LDL-induced MG53 reduction both in vitro and in vivo. Combined NAC and MG53 treatment significantly improved MAPC survival against ox-LDL. These data suggested that NAC enhanced the protective effect of MG53 on MAPCs against ox-LDL through preventing ox-LDL-induced reduction of MG53.


Subject(s)
Acetylcysteine/pharmacology , Bone Marrow Cells/drug effects , Gene Expression Regulation/drug effects , Lipoproteins, LDL/toxicity , Membrane Proteins/metabolism , Multipotent Stem Cells/drug effects , Protective Factors , Animals , Apoptosis , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Cycle , Cell Proliferation , Free Radical Scavengers/pharmacology , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/pathology , Rats
10.
J Cell Physiol ; 235(11): 8640-8652, 2020 11.
Article in English | MEDLINE | ID: mdl-32324269

ABSTRACT

We recently reported that epiblast stem cells (EpiSCs)-like cells could be derived from preimplantation embryos (named as AFSCs). Here, we established AFSCs from pre-implantation embryos of multiple mouse strains and showed that unlike EpiSCs, the derivation efficiency of AFSCs was affected by the genetic background. We then used AFSCs lines to dissect the roles of Activin A (Act A) and basic fibroblast growth factor and reported that Act A alone was capable of maintaining self-renewal but not developmental potential in vivo. Finally, we established a novel experimental system, in which AFSCs were efficiently converted to multipotent progenitor stem cells using Act A and bone morphogenetic protein 4 (named as ABSCs). Importantly, these ABSCs contributed to neural mesodermal progenitors and lateral plate mesoderm in postimplantation chimeras. Taken together, our study established a robust experimental system for the generation of specific multipotent progenitor stem cells that was self-renewable and capable of contributing to embryonic and extra-embryonic tissues.


Subject(s)
Activins/pharmacology , Germ Layers/drug effects , Multipotent Stem Cells/drug effects , Pluripotent Stem Cells/drug effects , Activins/metabolism , Animals , Blastocyst/drug effects , Blastocyst/metabolism , Bone Morphogenetic Protein 4/drug effects , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Embryonic Development/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Germ Layers/growth & development , Mice , Pluripotent Stem Cells/metabolism , Signal Transduction/drug effects
11.
Am J Physiol Renal Physiol ; 318(4): F861-F869, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32003597

ABSTRACT

Serum soluble Fas (sFas) levels are associated with erythropoietin (Epo) hyporesponsiveness in patients with chronic kidney disease (CKD). Whether sFas could predict the need for erythropoiesis-stimulating agent (ESA) usage and its influence in erythropoiesis remain unclear. We evaluated the relation between sFas and ESA therapy in patients with CKD with anemia and its effect on erythropoiesis in vitro. First, we performed a retrospective cohort study with 77 anemic patients with nondialysis CKD. We performed in vitro experiments to investigate whether sFas could interfere with the behavior of hematopoietic stem cells (HSCs). HSCs were isolated from umbilical cord blood and incubated with recombinant sFas protein in a dose-dependent manner. Serum sFas positively correlated with Epo levels (r = 0.30, P = 0.001) but negatively with hemoglobin (r = -0.55, P < 0.001) and glomerular filtration rate (r = -0.58, P < 0.001) in patients with CKD at baseline. Elevated sFas serum levels (4,316 ± 897 vs. 2,776 ± 749, P < 0.001) with lower estimated glomerular filtration rate (26.2 ± 10.1 vs. 33.5 ± 14.3, P = 0.01) and reduced hemoglobin concentration (11.1 ± 0.9 vs. 12.5 ± 1.2, P < 0.001) were identified in patients who required ESA therapy compared with patients with non-ESA. Afterward, we detected that the sFas level was slight correlated with a necessity of ESA therapy in patients with nondialysis CKD and anemia. In vitro assays demonstrated that the erythroid progenitor cell frequency negatively correlated with sFas concentration (r = -0.72, P < 0.001). There was decreased erythroid colony formation in vitro when CD34+ HSCs were incubated with a higher concentration of sFas protein (1.56 ± 0.29, 4.33 ± 0.53, P < 0.001). Our findings suggest that sFas is a potential predictor for ESA therapy in patients with nondialysis CKD and that elevated sFas could affect erythropoiesis in vitro.


Subject(s)
Anemia/blood , Erythropoiesis , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Renal Insufficiency, Chronic/complications , fas Receptor/blood , Adult , Aged , Anemia/diagnosis , Anemia/drug therapy , Anemia/etiology , Biomarkers/blood , Brazil , Cells, Cultured , Clinical Decision-Making , Databases, Factual , Erythropoiesis/drug effects , Erythropoietin/blood , Female , Hematinics/therapeutic use , Hematopoietic Stem Cells/drug effects , Hemoglobins/metabolism , Humans , Male , Middle Aged , Multipotent Stem Cells/drug effects , North Carolina , Patient Selection , Predictive Value of Tests , Recombinant Proteins/pharmacology , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/diagnosis , Retrospective Studies
12.
Alcohol Clin Exp Res ; 44(9): 1734-1746, 2020 09.
Article in English | MEDLINE | ID: mdl-32671866

ABSTRACT

BACKGROUND: Stem cells present in the vessel wall may be triggered in response to injurious stimuli to undergo differentiation and contribute to vascular disease development. Our aim was to determine the effect of moderate alcohol (EtOH) exposure on the expansion and differentiation of S100 calcium-binding protein B positive (S100ß+ ) resident vascular stem cells and their contribution to pathologic vessel remodeling in a mouse model of arteriosclerosis. METHODS AND RESULTS: Lineage tracing analysis of S100ß+ cells was performed in male and female S100ß-eGFP/Cre/ERT2-dTomato transgenic mice treated daily with or without EtOH by oral gavage (peak BAC: 15 mM or 0.07%) following left common carotid artery ligation for 14 days. Carotid arteries (ligated or sham-operated) were harvested for morphological analysis and confocal assessment of fluorescent-tagged S100 ß + cells in FFPE carotid cross sections. Ligation-induced carotid remodeling was more robust in males than in females. EtOH-gavaged mice had less adventitial thickening and markedly reduced neointimal formation compared to controls, with a more pronounced inhibitory effect in males compared to females. There was significant expansion of S100ß+ -marked cells in vessels postligation, primarily in the neointimal compartment. EtOH treatment reduced the fraction of S100ß+ cells in carotid cross sections, concomitant with attenuated remodeling. In vitro, EtOH attenuated Sonic Hedgehog-stimulated myogenic differentiation (as evidenced by reduced calponin and myosin heavy chain expression) of isolated murine S100ß+ vascular stem cells. CONCLUSIONS: These data highlight resident vascular S100ß+ stem cells as a novel target population for alcohol and suggest that regulation of these progenitors in adult arteries, particularly in males, may be an important mechanism contributing to the antiatherogenic effects of moderate alcohol consumption.


Subject(s)
Arteriosclerosis/pathology , Carotid Artery, Common/drug effects , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Multipotent Stem Cells/drug effects , S100 Calcium Binding Protein beta Subunit/metabolism , Vascular Remodeling/drug effects , Alcohol Drinking , Animals , Arteriosclerosis/metabolism , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Ligation , Mice , Mice, Transgenic , Microscopy, Confocal , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/pathology , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Neointima/metabolism , Neointima/pathology
13.
Cell Tissue Bank ; 21(4): 655-666, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32564258

ABSTRACT

Low survival rate of grafted mesenchymal stem cells (MSC) in injured tissue is one of the major limitations of stem cell therapy. One of the most important factors that limits the MSCs survival rate and retention is ischemic stress, which can lead to damage to all components of the cell. In particular, it can damage mitochondria, that play an important role in apoptosis with releasing apoptotic factors. Therefore, we investigated the protective effects of Acetyl-L-carnitine (ALCAR) against serum and glucose deprivation (SGD) in adipose-derived mesenchymal stem cells (AD-MSCs). We measured cell viability, proliferation, and apoptosis in cells experiencing SGD stress for 8 h with exposure to varying concentrations of ALCAR. Results showed that ALCAR protects cells against SGD stress by reducing apoptosis. Its protective effects are associated with reductions in cleaved caspase-3 and attenuation of apoptosis. Result showed that ALCAR exhibits protective effects against SGD-induced damage to AD-MSCs by enhancing the expression of survival signals and by decreasing the expression of death signals.


Subject(s)
Acetylcarnitine/pharmacology , Apoptosis/drug effects , Glucose/deficiency , Mesenchymal Stem Cells/cytology , Protective Agents/pharmacology , Animals , Caspase 3/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Culture Media, Serum-Free , DNA Fragmentation/drug effects , Male , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Rats, Wistar
14.
Bull Exp Biol Med ; 168(3): 356-360, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31938906

ABSTRACT

Peculiar roles of JAKs and STAT3 in realization of growth potential of various types of progenitor cells in neural tissue were examined during ethanol-induced neurodegeneration modeled both in vitro and in vivo. During in vitro action of C2H5OH, these signal molecules exerted the opposite effects on mitotic activity of multipotent neural stem cells and committed neural progenitors (the clonogenic PSA-NCAM+ cells). The JAKs and STAT3 inhibitors down-regulated the rate of neural stem cell division (proliferative activity) but up-regulated such activity of the committed neural progenitors. A long-term in vivo exposure of mice to ethanol inversed the roles of JAKs and STAT3 in determination of proliferative status of neural stem cells and eliminated involvement of JAKs in functional control over the committed progenitors of neurons. The data attest to much promise of STAT3 inhibitors in treatment of ethanol-induced CNS diseases as the remedies that stimulate realization of growth potential in multipotent neural stem cells and committed neural progenitors.


Subject(s)
Ethanol/toxicity , Janus Kinases/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Mice , Mice, Inbred C57BL , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Neural Stem Cells/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects
15.
J Cell Physiol ; 234(5): 7569-7578, 2019 05.
Article in English | MEDLINE | ID: mdl-30368818

ABSTRACT

Stem cells are often transplanted with scaffolds for tissue regeneration; however, how the mechanical property of a scaffold modulates stem cell fate in vivo is not well understood. Here we investigated how matrix stiffness modulates stem cell differentiation in a model of vascular graft transplantation. Multipotent neural crest stem cells (NCSCs) were differentiated from induced pluripotent stem cells, embedded in the hydrogel on the outer surface of nanofibrous polymer grafts, and implanted into rat carotid arteries by anastomosis. After 3 months, NCSCs differentiated into smooth muscle cells (SMCs) near the outer surface of the polymer grafts; in contrast, NCSCs differentiated into glial cells in the most part of the hydrogel. Atomic force microscopy demonstrated a stiffer matrix near the polymer surface but much lower stiffness away from the polymer graft. Consistently, in vitro studies confirmed that stiff surface induced SMC genes whereas soft surface induced glial genes. These results suggest that the scaffold's mechanical properties play an important role in directing stem cell differentiation in vivo, which has important implications in biomaterials design for stem cell delivery and tissue engineering.


Subject(s)
Cell Differentiation/physiology , Neural Crest/cytology , Neural Stem Cells/cytology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Humans , Hydrogels/pharmacology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Nanofibers/chemistry , Neural Crest/drug effects , Neural Stem Cells/drug effects , Neuroglia/cytology , Neuroglia/drug effects , Polymers/chemistry , Rats , Tissue Engineering/methods , Tissue Scaffolds
16.
Biochem Biophys Res Commun ; 518(1): 134-140, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31409482

ABSTRACT

The potential application of human induced pluripotent stem cells (hiPSCs) brings great expectations to regenerative medicine. However, several safety concerns, such as oncogenic transformation, remain. A number of methods have been developed to produce hiPSCs with potentially reduced risks. Cell-penetrating peptides (CPPs) are expected to improve the efficiency of nonviral reprogramming by delivering biologically active molecules into cells. Here, we show that the transfection of CPPs alone into normal adult human fibroblasts generated embryonic body (EB)-like cell clusters in the absence of reprogramming factors. The CPP-generated cell clusters were positive for a set of multipotency markers and differentiated into endodermal, ectodermal, and mesodermal cells in vitro. These results suggest that CPPs converted normal human adult somatic cells into multipotent cells. Moreover, we show that CPPs dissociated histone deacetylase 1 and lysine-specific demethylase 1 from the promoter/enhancer regions of reprogramming factors to reactivate their expression. This is the first report of an easy and quick method for somatic cell reprogramming by CPPs and a novel mechanism of reprogramming. The potential application of CPP-generated multipotent cells resolves several concerns, especially safety issues, in regenerative medicine.


Subject(s)
Cell Differentiation/drug effects , Cell-Penetrating Peptides/pharmacology , Fibroblasts/cytology , Multipotent Stem Cells/cytology , Amino Acid Sequence , Animals , Cell Aggregation/drug effects , Cell Line , Cell-Penetrating Peptides/chemistry , Embryoid Bodies/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Mice, Inbred NOD , Mice, SCID , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Mutant Proteins/pharmacology
17.
Int J Mol Sci ; 20(24)2019 Dec 11.
Article in English | MEDLINE | ID: mdl-31835689

ABSTRACT

There is a current clinical need for the development of bone void fillers and bioactive bone graft substitutes. The use of mesenchymal stem cells (MSCs) that are seeded into 3D scaffolds and induce bone generation in the event of MSCs osteogenic differentiation is highly promising. Since calcium ions and phosphates promote the osteogenic differentiation of MSCs, the use of the calcium complexes of phosphate-containing polymers is highly prospective in the development of osteogenic scaffolds. Calcium poly(ethylene phosphate)s (PEP-Ca) appear to be potentially suitable candidates primarily because of PEP's biodegradability. In a series of experiments with human adipose-tissue-derived multipotent mesenchymal stem cells (ADSCs), we demonstrated that PEP-Ca are non-toxic and give rise to osteogenesis gene marker, bone morphogenetic protein 2 (BMP-2) and mineralization of the intercellular matrix. Owing to the synthetic availability of poly(ethylene phosphoric acid) block copolymers, these results hold out the possibility for the development of promising new polymer composites for orthopaedic and maxillofacial surgery.


Subject(s)
Calcium Phosphates/pharmacology , Calcium/pharmacology , Cell Differentiation/drug effects , Mesenchymal Stem Cells/cytology , Osteogenesis/drug effects , Polyethylene/pharmacology , Calcification, Physiologic/drug effects , Calcium/chemistry , Calcium Phosphates/chemistry , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Phosphoric Acids/chemical synthesis , Phosphoric Acids/chemistry , Polyethylene/chemistry
18.
Bull Exp Biol Med ; 167(1): 182-188, 2019 May.
Article in English | MEDLINE | ID: mdl-31177456

ABSTRACT

We studied the effect of non-thermal argon plasma on proliferative activity of bone marrow multipotent stromal cells in vitro. Treatment of stromal cell suspension with pure argon did not affect their proliferation. The cells treated with non-thermal argon plasma and explanted in the treatment medium demonstrated growth inhibition by 30-40% in comparison with the control. Multipotent stromal cells treated with plasma and after centrifugation explanted in normal medium within 12 min demonstrated accelerated growth. The total cell growth from the pellet and supernatant significantly exceeded the control values. We also analyzed adhesion and proliferative activity of multipotent stromal cells treated with non-thermal plasma on bioresorbable carriers. The cells adhered and proliferated on all types of studied samples. Adhesion properties of scaffolds differed. Caprolactone was found to be the most suitable material for adhesion and proliferation of multipotent stromal cells.


Subject(s)
Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Plasma Gases/pharmacology , Tissue Engineering/methods , Animals , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Rabbits , Tissue Scaffolds/chemistry
19.
Bull Exp Biol Med ; 166(3): 348-352, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30627912

ABSTRACT

One hour after polyvinylpyrrolidone administration, the content of multipotent stromal cells in the spleen of CBA and CBA/N mice increased almost equally (by 2.5 and 2.9 times, respectively), but in 24 h, the effectiveness of multipotent stromal cell cloning in the spleen of CBA/N mice decreased almost to the control level, whereas in CBA mice, the number of multipotent stromal cells continued to increase. Serum concentration of IL-5, TNFα, and IL-2 in both lines was elevated in 1 h after polyvinylpyrrolidone administration, which is likely to reflect activation of the innate immunity. One day after polyvinylpyrrolidone administration, the number of multipotent stromal cells in bone marrow transplants in the CBA/N→CBA/N and CBA→CBA/N groups remained practically unchanged, while in groups CBA→CBA and CBA/N→CBA it was equally increased (by 3.6 and 3.4 times, respectively). Thus, the number of multipotent stromal cells in bone marrow transplants after 1 day was increased only in groups where recipients (CBA mice) were capable of responding to polyvinylpyrrolidone administration, i.e. the number of stromal cells by this term, was apparently determined by the presence of activated immunocompetent cells. These findings also indicate that activation of the stromal tissue dur ing immune response can have a two-phasic pattern: the first phase (1 h after antigen adminis tration) can be determined by activation of innate immunity receptors (in multipotent stromal cells or other cells) observed in CBA and CBA/N mice, and the second phase occurs during further development of the immune response (that was observed in CBA mice, but not in CBA/N mice due to absence of CD+B-1a lymphocytes). The findings attest to close interactions between the stromal tissue and the immune system.


Subject(s)
Bone Marrow Cells/drug effects , Cell Communication/drug effects , Multipotent Stem Cells/drug effects , Povidone/pharmacology , Vaccines, Synthetic/pharmacology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Cell Communication/immunology , Cell Count , Clone Cells , Host Specificity , Immunity, Innate/drug effects , Interleukin-2/blood , Interleukin-2/immunology , Interleukin-5/blood , Interleukin-5/immunology , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred CBA , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
20.
J Cell Mol Med ; 22(3): 1840-1854, 2018 03.
Article in English | MEDLINE | ID: mdl-29314633

ABSTRACT

The wound healing is a complex process wherein inflammation, proliferation and regeneration evolve according to a spatio-temporal pattern from the activation of coagulation cascade to the formation of a plug clot including fibrin matrix, blood-borne cells and cytokines/growth factors. Creating environments conducive to tissue repair, the haemoderivatives are commonly proposed for the treatment of hard-to-heal wounds. Here, we explored in vitro the intrinsic regenerative potentialities of a leucocyte- and platelet-rich fibrin product, known as CPL-MB, defining the stemness grade of cells sprouting from the haemoderivative. Using highly concentrated serum-based medium to simulate wound conditions, we isolated fibroblast-like cells (CPL-CMCs) adhering to plastic and showing stable in vitro propagation, heterogeneous stem cell expression pattern, endothelial adhesive properties and immunomodulatory profile. Due to their blood derivation and expression of CXCR4, CPL-CMCs have been suggested to be immature cells circulating in peripheral blood at quiescent state until activation by both coagulation event and inflammatory stimuli such as stromal-derived factor 1/SDF1. Expressing integrins (CD49f, CD103), vascular adhesion molecules (CD106, CD166), endoglin (CD105) and remodelling matrix enzymes (MMP2, MMP9, MMP13), they showed a transendothelial migratory potential besides multipotency. Taken together, our data suggested that a standardized, reliable and economically feasible blood product such as CPL-MB functions as an artificial stem cell niche that, under permissive conditions, originate ex vivo immature cells that could be useful for autologous stem cell-based therapies.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Leukocytes/metabolism , Multipotent Stem Cells/metabolism , Platelet-Rich Fibrin/metabolism , Regenerative Medicine/methods , Adipogenesis/drug effects , Adipogenesis/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling/methods , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Neurogenesis/drug effects , Neurogenesis/genetics , Proteomics/methods , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL