Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
Annu Rev Cell Dev Biol ; 30: 677-704, 2014.
Article in English | MEDLINE | ID: mdl-25150008

ABSTRACT

Two opposing descriptions of so-called mesenchymal stem cells (MSCs) exist at this time. One sees MSCs as the postnatal, self-renewing, and multipotent stem cells for the skeleton. This cell coincides with a specific type of bone marrow perivascular cell. In skeletal physiology, this skeletal stem cell is pivotal to the growth and lifelong turnover of bone and to its native regeneration capacity. In hematopoietic physiology, its role as a key player in maintaining hematopoietic stem cells in their niche and in regulating the hematopoietic microenvironment is emerging. In the alternative description, MSCs are ubiquitous in connective tissues and are defined by in vitro characteristics and by their use in therapy, which rests on their ability to modulate the function of host tissues rather than on stem cell properties. Here, I discuss how the two views developed, conceptually and experimentally, and attempt to clarify the confusion arising from their collision.


Subject(s)
Mesenchymal Stem Cells/cytology , Animals , Bone Marrow Cells/classification , Bone Marrow Cells/cytology , Bone and Bones/cytology , CD146 Antigen/analysis , Cell Separation/methods , Cell- and Tissue-Based Therapy , Cells, Cultured , Clone Cells/cytology , Connective Tissue/immunology , Humans , Immunomodulation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/classification , Mice , Models, Biological , Pericytes/cytology , Pluripotent Stem Cells/cytology , Radiation Chimera , Stem Cell Niche , Stromal Cells/classification , Stromal Cells/cytology , Transplantation, Heterotopic
2.
Cytometry A ; 95(7): 769-781, 2019 07.
Article in English | MEDLINE | ID: mdl-30861637

ABSTRACT

Mass cytometry by time-of-flight (CyTOF) is a valuable technology for high-dimensional analysis at the single cell level. Identification of different cell populations is an important task during the data analysis. Many clustering tools can perform this task, which is essential to identify "new" cell populations in explorative experiments. However, relying on clustering is laborious since it often involves manual annotation, which significantly limits the reproducibility of identifying cell-populations across different samples. The latter is particularly important in studies comparing different conditions, for example in cohort studies. Learning cell populations from an annotated set of cells solves these problems. However, currently available methods for automatic cell population identification are either complex, dependent on prior biological knowledge about the populations during the learning process, or can only identify canonical cell populations. We propose to use a linear discriminant analysis (LDA) classifier to automatically identify cell populations in CyTOF data. LDA outperforms two state-of-the-art algorithms on four benchmark datasets. Compared to more complex classifiers, LDA has substantial advantages with respect to the interpretable performance, reproducibility, and scalability to larger datasets with deeper annotations. We apply LDA to a dataset of ~3.5 million cells representing 57 cell populations in the Human Mucosal Immune System. LDA has high performance on abundant cell populations as well as the majority of rare cell populations, and provides accurate estimates of cell population frequencies. Further incorporating a rejection option, based on the estimated posterior probabilities, allows LDA to identify previously unknown (new) cell populations that were not encountered during training. Altogether, reproducible prediction of cell population compositions using LDA opens up possibilities to analyze large cohort studies based on CyTOF data. Ā© 2019 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.


Subject(s)
Bone Marrow Cells/classification , Flow Cytometry/methods , Single-Cell Analysis/methods , Algorithms , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cluster Analysis , Datasets as Topic , Humans , Mice , Reproducibility of Results
3.
J Med Syst ; 43(4): 82, 2019 Feb 23.
Article in English | MEDLINE | ID: mdl-30798374

ABSTRACT

In the detection of myeloproliferative, the number of cells in each type of bone marrow cells (BMC) is an important parameter for the evaluation. In this study, we propose a new counting method, which consists of three modules including localization, segmentation and classification. The localization of BMC is achieved from a color transformation enhanced BMC sample image and stepwise averaging method. In the nucleus segmentation, both stepwise averaging method and Otsu's method are applied to obtain a weighted threshold for segmenting the patch into nucleus and non-nucleus. In the cytoplasm segmentation, a color weakening transformation, an improved region growing method and the K-Means algorithm are employed. The connected cells with BMC will be separated by the marker-controlled watershed algorithm. The features will be extracted for the classification after the segmentation. In this study, the BMC are classified using the support vector machine into five classes; namely, neutrophilic split granulocyte, neutrophilic stab granulocyte, metarubricyte, mature lymphocytes and the outlier (all other cells not listed). Experimental results show that the proposed method achieves superior segmentation and classification performance with an average segmentation accuracy of 91.76% and an average recall rate of 87.49%. The comparison shows that the proposed segmentation and classification methods outperform the existing methods.


Subject(s)
Bone Marrow Cells/classification , Image Processing, Computer-Assisted/methods , Microscopy/methods , Pattern Recognition, Automated/methods , Support Vector Machine , Algorithms , Cell Nucleus , Color , Humans
4.
J Cell Physiol ; 233(12): 9739-9749, 2018 12.
Article in English | MEDLINE | ID: mdl-29987913

ABSTRACT

Bone-marrow-derived mesenchymal stem cells (MSCs) have great potential in transplantation medicine due to their multiple advantages. However, the controlled differentiation of MSCs is one of the key aspects of effective clinical transplantation. Growing evidence suggests that the cell cycle plays an important role in regulating differentiation, while p130 and E2F4 are key to cell cycle checkpoints. The aim of the study is to evaluate the effects and mechanism of p130/E2F4 on the multidifferentiation of MSCs. Our data showed that the transduction efficiencies of p130 or E2F4 mediated by lentiviral vectors were 80.3%-84.4%. p130 and E2F4 mRNA expression was significantly higher in MSC-p130 and MSC-E2F4 cells than in MSC normal control (NC) cells. Similar results were also observed for p130 and E2F4 protein expression. After osteogenic or adipogenic differentiation, the G1 phase was significantly delayed in the MSC-p130 and MSC-E2F4 groups compared with that in the MSC-NC group. However, the G1 phase in the MSC-p130 and MSC-E2F4 groups did the opposite after chondrogenic differentiation. Moreover, overexpressing p130 or E2F4 significantly improved osteogenic differentiation while inhibiting adipogenic and chondrogenic differentiation of mouse MSCs (mMSCs). Moreover, overexpressing p130 or E2F4 significantly improved migration but not proliferation of mMSCs. Our data suggest that cell cycle regulation may be involved in p130/E2F4-mediated changes in the multipotential abilities of bone-marrow-derived mMSCs.


Subject(s)
Cell Differentiation/genetics , Crk-Associated Substrate Protein/genetics , E2F4 Transcription Factor/genetics , Mesenchymal Stem Cells/metabolism , Adipogenesis/genetics , Bone Marrow Cells/classification , Bone Marrow Cells/metabolism , Cell Cycle Checkpoints/genetics , Cell Movement/genetics , Gene Expression Regulation, Developmental/genetics , Genetic Vectors , Humans , Lentivirus/genetics , Mesenchymal Stem Cells/cytology , Osteogenesis/genetics
5.
Methods ; 112: 68-74, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27582124

ABSTRACT

Adult humans need to make 2.5million red blood cells (RBCs) every second to maintain a steady state level of 25trillion circulating RBCs. Understanding normal erythropoiesis as well as diseases that afflict the erythron, such as genetic anemias, hyperproliferative disorders, and myelodysplastic syndromes, requires a robust method to delineate erythropoietic intermediates. In order to apply the power of flow cytometry to these studies, challenges of limited immunophenotypic markers, incorporation of significant changes in morphology, and maturational changes that occur along a continuum need to be met. Imaging flow cytometry (IFC) provides a solution to address these challenges. Integration of changes in immunophenotype, loss of RNA (ribosomes), and enucleation, with morphological characteristics of cell and nuclear size, can be used to delineate erythroblasts that correlate with classical histological classifications. A protocol is described that demonstrates the basic approaches of staining panel selection, mask generation and selection of features to best sequentially refine erythroid intermediates and remove contaminating cells with overlapping immunophenotype. Ultimately erythroid cells in the murine bone marrow are divided into seven sub-populations using IFC including four erythroblasts (pro-, basophilic, polychromatophilic and orthochromatic), the pyrenocyte, which contains the eliminated nucleus, the enucleated reticulocyte and the mature RBC.


Subject(s)
Bone Marrow Cells/cytology , Cell Lineage/genetics , Erythropoiesis/genetics , Flow Cytometry/methods , Image Cytometry/methods , Animals , Biomarkers/metabolism , Bone Marrow Cells/classification , Bone Marrow Cells/metabolism , Cell Cycle/genetics , Cell Differentiation , Cell Nucleus/ultrastructure , Erythroblasts/cytology , Erythroblasts/metabolism , Erythrocytes/cytology , Erythrocytes/metabolism , Flow Cytometry/instrumentation , Humans , Image Cytometry/instrumentation , Mice , Primary Cell Culture , Reticulocytes/cytology , Reticulocytes/metabolism , Ribosomes/ultrastructure , Staining and Labeling/methods
6.
J Clin Lab Anal ; 32(1)2018 Jan.
Article in English | MEDLINE | ID: mdl-28220972

ABSTRACT

BACKGROUND: Morphological characteristics of blood cells are still qualitatively defined. So a texture analysis (Tx) method using gray level co-occurrence matrices (GLCMs; CM-Tx method) was applied to images of erythrocyte precursor cells (EPCs) for quantitatively distinguishing four types of EPC stages: proerythroblast, basophilic erythroblast, polychromatic erythroblast, and orthochromatic erythroblast. METHODS: Fifty-five images of four types of EPCs were downloaded from an atlas uploaded by the Blood Cell Morphology Standardization Subcommittee (BCMSS) of the Japanese Society of Laboratory Hematology (JSLH). Using in-house programs, two types of GLCMs-(R: d=1, ƎĀø=0Ā°) and (U: d=1, ƎĀø=270Ā°)-and nine types of texture distinction index (TDI) were calculated with images removed outer part of cell. RESULTS: Three binary decision trees were sequentially divided among four types of EPC with the sum average of GLCM (U), the contrast of GLCM (R), and the sum average of GLCM (U). The average concordance rate (sensitivity) of CM-Tx method with the judgments of eleven experts in the BCMSS of the JSLH was 95.8% (87.5-100.0), and the average specificity was 97.6% (92.5-100.0). CONCLUSIONS: The CM-Tx method is an effective tool for quantitative distinction of EPC with their morphological features.


Subject(s)
Blood Cells/cytology , Bone Marrow Cells/cytology , Cytological Techniques/methods , Image Processing, Computer-Assisted/methods , Blood Cells/classification , Bone Marrow Cells/classification , Humans , Microscopy
7.
Blood ; 125(11): 1739-48, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25573986

ABSTRACT

Specific serum antibodies mediating humoral immunity and autoimmunity are provided by mature plasma cells (PC) residing in the bone marrow (BM), yet their dynamics and composition are largely unclear. We here characterize distinct subsets of human PC differing by CD19 expression. Unlike CD19(+) PC, CD19(-) PC were restricted to BM, expressed predominantly IgG, and they carried a prosurvival, distinctly mature phenotype, that is, HLA-DR(low)Ki-67(-)CD95(low)CD28(+)CD56(+/-), with increased BCL2 and they resisted their mobilization from the BM after systemic vaccination. Fewer mutations within immunoglobulin VH rearrangements of CD19(-) BMPC may indicate their differentiation in early life. Their resistance to in vivo B-cell depletion, that is, their independency from supply with new plasmablasts, is consistent with long-term stability of this PC subset in the BM. Moreover, CD19(-) PC were detectable in chronically inflamed tissues and secreted autoantibodies. We propose a multilayer model of PC memory in which CD19(+) and CD19(-) PC represent dynamic and static components, respectively, permitting both adaptation and stability of humoral immune protection.


Subject(s)
Antigens, CD19/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Immunoglobulin G/metabolism , Plasma Cells/immunology , Antilymphocyte Serum/administration & dosage , Bone Marrow Cells/classification , Boronic Acids/administration & dosage , Bortezomib , Cell Differentiation , Cell Survival , Diphtheria-Tetanus Vaccine/administration & dosage , Humans , Immunity, Humoral , Immunologic Memory , Inflammation/immunology , Inflammation/pathology , Lymphocyte Depletion , Models, Immunological , Mutation , Phenotype , Plasma Cells/classification , Plasma Cells/cytology , Pyrazines/administration & dosage , V(D)J Recombination
8.
Arterioscler Thromb Vasc Biol ; 36(9): 1722-33, 2016 09.
Article in English | MEDLINE | ID: mdl-27386937

ABSTRACT

Monocytes and macrophages are key immune cells involved in the early progression of atherosclerosis. Transcription factors that control their development in the bone marrow are important therapeutic targets to control the numbers and functions of these cells in disease. This review highlights what is currently known about the transcription factors that are critical for monocyte development.


Subject(s)
Bone Marrow Cells/physiology , Cell Differentiation , Monocytes/physiology , Transcription, Genetic , Animals , Bone Marrow Cells/classification , Bone Marrow Cells/immunology , Cellular Microenvironment , Gene Expression Regulation , Genotype , Hematopoiesis, Extramedullary , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Monocytes/classification , Monocytes/immunology , Myelopoiesis , Phenotype , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Blood ; 121(1): 11-9, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23053574

ABSTRACT

The developmental origin of IFN-producing plasmacytoid dendritic cells (pDCs) has been uncertain. In the present study, we tracked the development of pDCs in cultures of BM precursors stimulated with Flt3 ligand. Common myeloid precursors (CMPs) produced both conventional DCs (cDCs) and pDCs via the DC-restricted common DC precursor. Common lymphoid precursors (CLPs) produced only a few cDCs with variable efficiency, but produced pDCs via a transient intermediate precursor with B-cell potential. The pDCs of both origins produced IFN-α when stimulated with CpG oligonucleotides. The pDCs of CLP origin showed evidence of past RAG1 expression and had D-J rearrangements in IgH genes. Most pDCs and all cDCs of CMP origin lacked these signs of a lymphoid past. However, in these cultures, some pDCs of CMP origin showed evidence of past RAG1 expression and had D-J IgH gene rearrangements; most of these derived from a subset of CMPs already expressing RAG1.


Subject(s)
Dendritic Cells/cytology , Lymphopoiesis/physiology , Myelopoiesis/physiology , Adoptive Transfer , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Bone Marrow Cells/classification , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Lineage , Cell Separation , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Clone Cells/cytology , Clone Cells/metabolism , CpG Islands , Dendritic Cells/metabolism , Gene Expression Regulation, Developmental , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Immunophenotyping , Interferon-alpha/biosynthesis , Interferon-alpha/genetics , Lymphopoiesis/genetics , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Myelopoiesis/genetics , Oligonucleotides/pharmacology , Radiation Chimera , Specific Pathogen-Free Organisms
10.
J Immunol ; 188(7): 3208-16, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22351937

ABSTRACT

The earliest thymic progenitors (ETPs) were recently shown to give rise to both lymphoid and myeloid cells. Whereas the majority of ETPs are derived from IL-7Rα-positive cells and give rise exclusively to T cells, the origin of the myeloid cells remains undefined. In this study, we show both in vitro and in vivo that IL-13Rα1(+) ETPs yield myeloid cells with no potential for maturation into T cells, whereas IL-13Rα1(-) ETPs lack myeloid potential. Moreover, transfer of lineage-negative IL-13Rα1(+) bone marrow stem cells into IL-13Rα1-deficient mice reconstituted thymic IL-13Rα1(+) myeloid ETPs. Myeloid cells or macrophages in the thymus are regarded as phagocytic cells whose function is to clear apoptotic debris generated during T cell development. However, the myeloid cells derived from IL-13Rα1(+) ETPs were found to perform Ag-presenting functions. Thus, IL-13Rα1 defines a new class of myeloid restricted ETPs yielding APCs that could contribute to development of T cells and the control of immunity and autoimmunity.


Subject(s)
Antigen-Presenting Cells/cytology , Antigens, Differentiation/analysis , Bone Marrow Cells/classification , Granulocyte-Macrophage Progenitor Cells/cytology , Interleukin-13 Receptor alpha1 Subunit/analysis , Myelopoiesis , Thymus Gland/cytology , Animals , Antigen-Presenting Cells/chemistry , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Bone Marrow Cells/chemistry , Cell Lineage , Cell Movement , Cells, Cultured , Female , Gene Knock-In Techniques , Granulocyte-Macrophage Progenitor Cells/chemistry , Granulocyte-Macrophage Progenitor Cells/drug effects , Granulocyte-Macrophage Progenitor Cells/immunology , Interleukin-13/pharmacology , Interleukin-13 Receptor alpha1 Subunit/deficiency , Interleukin-13 Receptor alpha1 Subunit/genetics , Lymphocytes, Null/cytology , Lymphopoiesis , Male , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Sequence Deletion , T-Lymphocytes/cytology
11.
Blood ; 117(17): 4467-75, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21364193

ABSTRACT

During development in the bone marrow (BM), NK-cell positioning within specific niches can be influenced by expression of chemokine or adhesion receptors. We previously demonstrated that the maintenance in the BM of selected NK-cell subsets is regulated by the CXCR4/CXCL12 axis. In the present study, we showed that CX3CR1 is prevalently expressed on KLRG1(+) NK cells, a subset considered terminally differentiated. Two KLRG1(+) NK-cell populations endowed with distinct homing and functional features were defined according to CX3CR1 expression. In the BM, KLRG1(+)/CX3CR1(-) NK cells were mainly positioned into parenchyma, while KLRG1(+)/CX3CR1(+) NK cells exhibited reduced CXCR4 expression and were preferentially localized in the sinusoids. We also showed that α(4) integrin plays a pivotal role in the maintenance of NK cells in the BM sinusoids and that α(4) neutralization leads to strong reduction of BM KLRG1(+)/CX3CR1(+) NK cells. Moreover, we found that KLRG1(+)/CX3CR1(+) cells originate from KLRG1(+)/CX3CR1(-) NK-cell population and display impaired capability to produce IFN-ƎĀ³ and to lyse YAC-1 target cells on cytokine stimulation. Altogether, our findings show that CX3CR1 represents a marker of a KLRG1(+) NK-cell population with unique properties that can irreversibly differentiate from the KLRG1(+)/CX3CR1(-) NK cells during steady state conditions.


Subject(s)
Biomarkers , Bone Marrow Cells/physiology , Killer Cells, Natural/physiology , Receptors, Chemokine/genetics , Receptors, Immunologic/genetics , Animals , Bone Marrow Cells/classification , Bone Marrow Cells/cytology , CX3C Chemokine Receptor 1 , Cell Differentiation/immunology , Chemokine CXCL12/metabolism , Female , Flow Cytometry , Gene Expression/immunology , Green Fluorescent Proteins/genetics , Integrin alpha4/metabolism , Killer Cells, Natural/classification , Killer Cells, Natural/cytology , Lectins, C-Type , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism
12.
Ann Hematol ; 92(5): 587-94, 2013 May.
Article in English | MEDLINE | ID: mdl-23307597

ABSTRACT

Laboratory mice play an outstanding role in modeling human development and disease. In contrast to human leukemia, the spleen is involved in almost all cases, and the bone marrow is only variably involved in murine models. Although mice have been used for medical research for over 100Ā years, there are only few reports with a small number of cases looking at morphology and quantitative composition of murine hematopoietic cells in the bone marrow of non-transplanted animals of most strains. To our knowledge, there is not even a single report describing the splenogram in C57BL/6J mice, one of the most commonly used strains for medical research. The present study illustrates the morphology of the hematopoietic cells in the bone marrow and spleen of non-treated C57BL/6J mice and establishes the murine myelogram from the largest healthy C57BL/6J cohort reported to date. Furthermore, we present the first murine splenogram described for C57BL/6J mice. Our study supports the acceptance of the presence of >5Ā % blast cells as providing clear evidence of abnormality in bone marrow like in humans. In addition, we are the first to show <1Ā % blast cells in the normal spleen. Interestingly, classical dysplastic changes were rare in normal healthy mice. Our study of the bone marrow and spleen of healthy non-transplanted animals provides reference ranges of each cell type and for the myeloid/erythroid ratio, which can be used to interpret preclinical gene therapy data, leukemogenesis, and hematopoiesis studies, and may improve the quality of such analyses.


Subject(s)
Bone Marrow Cells/cytology , Hematopoietic Stem Cells/cytology , Spleen/cytology , Animals , Bone Marrow/physiology , Bone Marrow Cells/classification , Cell Count , Cell Shape , Granulocyte Precursor Cells/cytology , Granulocyte Precursor Cells/physiology , Granulocytes/cytology , Granulocytes/physiology , Health , Hematopoiesis/physiology , Hematopoietic Stem Cells/classification , Macrophages/cytology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/physiology
13.
Bull Exp Biol Med ; 155(5): 601-4, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24288718

ABSTRACT

We studied the cell composition and free radical oxidation in the bone marrow of white outbred rats after hypokinetic stress (24 and 72 h) and the effects of exogenous glucocorticoid triamcinolone acetonide (2 mg/kg; injected 24 h after hypokinesia); the measurements were performed in 96 h after drug administration. The hypoplastic effect of the glucocorticoid after 24-h hypokinesia was observed against the background of reduced free radical oxidation. In animals subjected to 3-day hypokinesia, the resistance of the bone marrow to the hypoplastic effect of this drug was accompanied by activation of free radical processes.


Subject(s)
Bone Marrow Cells/drug effects , Glucocorticoids/pharmacology , Stress, Physiological , Triamcinolone Acetonide/pharmacology , Animals , Animals, Outbred Strains , Bone Marrow Cells/classification , Bone Marrow Cells/cytology , Free Radicals/metabolism , Immobilization , Lipid Peroxidation/drug effects , Male , Protein Carbonylation/drug effects , Rats
15.
J Oral Maxillofac Surg ; 70(8): e469-76, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22793961

ABSTRACT

PURPOSE: As part of the authors' research on potential osteogenesis by filling bone defects with human bone marrow-derived mesenchymal stromal cells (hBM-MSCs) in patients with cleft lip and palate, they examined the cytoproliferative potential and cytobiological activity of hBM-MSCs in vitro and their osteogenic potential in vivo without performing osteoinduction. MATERIALS AND METHODS: The hBM-MSCs were collected from iliac cancellous bone and then used in primary culture, followed by 2 subcultures using an autologous serum (AS)-containing medium and a fetal bovine serum (FBS)-containing medium. Cytoproliferative potential and cytobiological activity as expressed by bone markers (alkaline phosphatase and osteocalcin) in hBM-MSCs cultured in the AS-containing medium (AS-cultured hBM-MSCs) and the FBS-containing medium (FBS-cultured hBM-MSCs) were examined in vitro, and the osteogenic potential of AS- and FBS-cultured hBM-MSCs was examined in mice. RESULTS: On day 6 of the second subculture, the number of hBM-MSCs per milliliter of specimen from 8 pediatric patients was significantly larger (P < .05) in FBS-cultured compared with AS-cultured hBM-MSCs. The alkaline phosphatase activity of hBM-MSCs was significantly greater (P < .05) when cultured in the AS-containing medium compared with the FBS-containing medium. The in vivo study showed the formation of an osteoid-like matrix rather than definite bone tissue. CONCLUSIONS: 1) FBS is appropriate for the cytoproliferation of hBM-MSCs; 2) the AS-containing medium is likely to have a good possibility of inducing the differentiation of hBM-MSCs; and 3) AS-cultured hBM-MSCs contain a group of cells that spontaneously differentiate into an osteoid-like matrix without performing osteoinduction.


Subject(s)
Bone Marrow Cells/physiology , Mesenchymal Stem Cells/physiology , Osteogenesis/physiology , Alkaline Phosphatase/analysis , Animals , Biomarkers/analysis , Blood , Bone Marrow Cells/classification , Bone Matrix/cytology , Bone Matrix/physiology , Cell Count , Cell Culture Techniques , Cell Proliferation , Child , Culture Media , Durapatite , Female , Humans , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/classification , Mice , Mice, Nude , Mitochondria/classification , Osteocalcin/analysis , Subcutaneous Tissue/surgery , Tissue Scaffolds
16.
J Cell Biochem ; 112(7): 1713-21, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21416499

ABSTRACT

The last decade was dominated by dissemination of the notion that postnatal "mesenchymal stem cells," found primarily in bone marrow but also in other tissues, can generate multiple skeletal and nonskeletal tissues, and thus can be exploited to regenerate a broad range of tissues and organs. The concept of "mesenchymal stem cells" and its applicative implications represent a significant departure from the solidly proven notion that skeletal stem cells are found in the bone marrow (and not in other tissues). Recent data that sharpen our understanding of the identity, nature, origin, and in vivo function of the archetypal "mesenchymal stem cells" (bone marrow skeletal stem cells) point to their microvascular location, mural cell identity, and function as organizers and regulators of the hematopoietic microenvironment/niche. These advances bring back the original concept from which the notion of "mesenchymal stem cells" evolved, and clarify a great deal of experimental data that accumulated in the past decade. As a novel paradigm emerges that accounts for many facets of the biology of skeletal stem cells, a novel paradigm independently emerges for their applicative/translational use. The two paradigms meet each other back in the future.


Subject(s)
Bone Marrow Cells/cytology , Mesenchymal Stem Cells/cytology , Animals , Bone Marrow Cells/classification , Bone Regeneration , Humans , Mesenchymal Stem Cells/classification , Microvessels/cytology , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Neovascularization, Physiologic , Osteogenesis , Pericytes/classification , Pericytes/cytology , Regeneration , Regenerative Medicine/trends
17.
Semin Diagn Pathol ; 28(4): 273-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22195405

ABSTRACT

Multiparameter flow cytometry (FCM) is an excellent method to follow the expression patterns of differentiation antigens using monoclonal antibodies to surface and cytoplasmic proteins. Although several authors described various aberrant immunophenotypic features in the bone marrow of patients with myelodysplastic syndromes (MDS), the World Health Organization 2008 classification recommended that, only if 3 or more phenotypic abnormalities are found involving 1 or more of the myeloid lineages can the aberrant FCM findings be considered suggestive of MDS. In the absence of conclusive morphologic and/or cytogenetic features, FCM abnormalities alone were considered not sufficient to establish MDS diagnosis and further follow-up of the patients was recommended. Review of the literature gives accumulating evidence that FCM has become an important part of the integrated diagnostic work-up of patients with suspected MDS. Several studies have also reported FCM findings significant for prognosis and therapy choice in MDS patients. Technical progress in multicolor FCM and new analysis programs, together with ongoing efforts to standardize the methodology, will make it possible to apply FCM in individual risk assessment and choice of best therapy for MDS patients.


Subject(s)
Cell Separation/methods , Flow Cytometry/methods , Immunophenotyping/methods , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/diagnosis , Bone Marrow Cells/classification , Humans , World Health Organization
18.
Int J Biol Sci ; 17(15): 4192-4206, 2021.
Article in English | MEDLINE | ID: mdl-34803492

ABSTRACT

Bone marrow-derived mesenchymal stem cells (BM-MSCs) are multipotent stromal cells that have a critical role in the maintenance of skeletal tissues such as bone, cartilage, and the fat in bone marrow. In addition to providing microenvironmental support for hematopoietic processes, BM-MSCs can differentiate into various mesodermal lineages including osteoblast/osteocyte, chondrocyte, and adipocyte that are crucial for bone metabolism. While BM-MSCs have high cell-to-cell heterogeneity in gene expression, the cell subtypes that contribute to this heterogeneity in vivo in humans have not been characterized. To investigate the transcriptional diversity of BM-MSCs, we applied single-cell RNA sequencing (scRNA-seq) on freshly isolated CD271+ BM-derived mononuclear cells (BM-MNCs) from two human subjects. We successfully identified LEPRhiCD45low BM-MSCs within the CD271+ BM-MNC population, and further codified the BM-MSCs into distinct subpopulations corresponding to the osteogenic, chondrogenic, and adipogenic differentiation trajectories, as well as terminal-stage quiescent cells. Biological functional annotations of the transcriptomes suggest that osteoblast precursors induce angiogenesis coupled with osteogenesis, and chondrocyte precursors have the potential to differentiate into myocytes. We also discovered transcripts for several clusters of differentiation (CD) markers that were either highly expressed (e.g., CD167b, CD91, CD130 and CD118) or absent (e.g., CD74, CD217, CD148 and CD68) in BM-MSCs, representing potential novel markers for human BM-MSC purification. This study is the first systematic in vivo dissection of human BM-MSCs cell subtypes at the single-cell resolution, revealing an insight into the extent of their cellular heterogeneity and roles in maintaining bone homeostasis.


Subject(s)
Bone Marrow Cells/classification , Mesenchymal Stem Cells/metabolism , Sequence Analysis, RNA , Single-Cell Analysis/methods , Aged , Aged, 80 and over , Animals , Bone Density , Bone Marrow Cells/metabolism , CD56 Antigen/genetics , CD56 Antigen/metabolism , Cell Differentiation , Chondrocytes/physiology , Cluster Analysis , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Muscle Development , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism
19.
Vet Immunol Immunopathol ; 234: 110215, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33676089

ABSTRACT

Dendritic cells (DCs) are the most potent antigen presenting cells (APCs). Because of the difficulty in obtaining these cells directly from tissues, different sources of DCs are frequently used for in vitro experimentation and many of their biological and functional characteristics were studied using these systems. Until recently, it was assumed that specific culture conditions polarized the differentiation of either DCs or macrophages (Macs); however, it was shown that some DC culture systems in other species generate heterogeneous cell populations that can be identified according to their CD11c and MHC class II (MHC-II) expression. Following this approach, porcine DCs were directly isolated from peripheral blood or differentiated in vitro by culturing bone marrow (BM) progenitor cells or blood monocytes treated with growth factors. Mostly homogeneous monocyte-derived DCs (MoDCs) were obtained with similar phenotype and phagocytic characteristics to that of blood DCs. On the contrary, BM-derived DC (BMDC) cultures generated two distinct heterogeneous populations identified as MHC-II+ and MHC-II++ cells. BMDCs MHC-II+ had similar phenotypic and phagocytic characteristics to those of MoDCs and blood DCs. However, BMDCs MHC-II++ population expressed a higher amount of surface markers and transcribed genes associated with Macs-lineage exhibiting a higher phagocytic capacity than all the other cells. Noteworthy, every cell system expressed different genetic signatures. These results will help interpreting and re-interpreting data obtained using in vitro systems.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Age Factors , Animals , Antigen-Presenting Cells/classification , Bone Marrow Cells/classification , Cells, Cultured , Dendritic Cells/classification , Dendritic Cells/immunology , Dendritic Cells/physiology , Female , Flow Cytometry , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation , Macrophages/immunology , Macrophages/physiology , Male , Monocytes/immunology , Swine
20.
J Exp Med ; 189(4): 729-34, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9989988

ABSTRACT

Allogeneic and autologous marrow transplants are routinely used to correct a wide variety of diseases. In addition, autologous marrow transplants potentially provide opportune means of delivering genes in transfected, engrafting stem cells. However, relatively little is known about the mechanisms of engraftment in transplant recipients, especially in the nonablated setting and with regard to cells not of hemopoietic origin. In particular, this includes stromal cells and progenitors of the osteoblastic lineage. We have demonstrated for the first time that a whole bone marrow transplant contains cells that engraft and become competent osteoblasts capable of producing bone matrix. This was done at the individual cell level in situ, with significant numbers of donor cells being detected by fluorescence in situ hybridization in whole femoral sections. Engrafted cells were functionally active as osteoblasts producing bone before being encapsulated within the bone lacunae and terminally differentiating into osteocytes. Transplanted cells were also detected as flattened bone lining cells on the periosteal bone surface.


Subject(s)
Bone Marrow Cells/classification , Bone Marrow Transplantation , Chimera , Osteoblasts/transplantation , Osteogenesis , Animals , Female , Graft Survival , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred BALB C , Stromal Cells/transplantation , Y Chromosome
SELECTION OF CITATIONS
SEARCH DETAIL