Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Eur Cell Mater ; 42: 401-414, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34825700

ABSTRACT

Mesenchymal stem cells (MSCs) are promising cells for regenerative medicine therapies because they can differentiate towards multiple cell lineages. However, the occurrence of cellular senescence and the acquiring of the senescence-associated secretory phenotype (SASP) limit their clinical use. Since the transcription factor TWIST1 influences expansion of MSCs, its role in regulating cellular senescence was investigated. The present study demonstrated that silencing of TWIST1 in MSCs increased the occurrence of senescence, characterised by a SASP profile different from irradiation-induced senescent MSCs. Knowing that senescence alters cellular metabolism, cellular bioenergetics was monitored by using the Seahorse XF apparatus. Both TWIST1-silencing-induced and irradiation-induced senescent MSCs had a higher oxygen consumption rate compared to control MSCs, while TWIST1-silencing-induced senescent MSCs had a low extracellular acidification rate compared to irradiation-induced senescent MSCs. Overall, data indicated how TWIST1 regulation influenced senescence in MSCs and that TWIST1 silencing-induced senescence was characterised by a specific SASP profile and metabolic state.


Subject(s)
Mesenchymal Stem Cells , Senescence-Associated Secretory Phenotype , Cellular Senescence , Energy Metabolism , Gene Expression Regulation
2.
Osteoarthritis Cartilage ; 27(12): 1831-1840, 2019 12.
Article in English | MEDLINE | ID: mdl-31536814

ABSTRACT

OBJECTIVE: Synovium contains multipotent progenitor/stromal cells (MPCs) with potential to participate in cartilage repair. Understanding the identity of these MPCs will allow their therapeutic potential to be fully exploited. Hence this study aimed to identify primary synovial MPCs and characterize them in the context of cartilage regeneration. METHODS: Primary MPC/MPC-subset specific markers in synovium were identified by FACS analysis of uncultured cells. MPC-subsets from human synovium obtained from patients undergoing total knee arthroplasty were FACS sorted, cultured, immunophenotyped and chondrogenically differentiated. The anatomical localization of MPCs in synovium was examined using immunohistochemistry. Finally, the presence of these MPC subsets in healthy synovium obtained from human organ donors was examined. RESULTS: A combination of CD45, CD31, CD73 and CD90 can isolate two distinct MPC-subsets in synovium. These MPC-subsets, freshly isolated from synovium, did not express CD45 or CD31, but expressed CD73. Additionally, a sub-population of CD73+ cells also expressed CD90. CD45-CD31-CD73+CD90- cells were significantly more chondrogenic than CD45-CD31-CD73+CD90+ cells in the presence of TGFß1. Interestingly, reduced chondrogenic ability of CD73+CD90+ cells could be reversed by the addition of BMP2, showing discrete chondrogenic factor requirements by distinct cell-subsets. In addition, these MPCs had distinct anatomical localization; CD73 was expressed both in intimal and sub-intimal region while CD90 was enriched in the sub-intimal region. We further demonstrated that these subsets are also present in healthy synovium. CONCLUSIONS: We provide indications that primary MPCs in synovial intima and sub-intima are phenotypically and functionally distinct with different chondrogenic properties.


Subject(s)
Cartilage, Articular/physiology , Cell Differentiation/physiology , Chondrogenesis/physiology , Multipotent Stem Cells/metabolism , Osteoarthritis, Knee , Regeneration/physiology , 5'-Nucleotidase/metabolism , Aged , Aged, 80 and over , Case-Control Studies , Cell Adhesion Molecules/metabolism , Female , Flow Cytometry , GPI-Linked Proteins/metabolism , Humans , Immunohistochemistry , Immunophenotyping , Leukocyte Common Antigens/metabolism , Male , Middle Aged , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, Chemokine/metabolism , Receptors, Growth Factor/metabolism , Synovial Membrane/cytology , Thy-1 Antigens/metabolism
3.
Biofabrication ; 11(3): 035012, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30921774

ABSTRACT

Decellularized tissue matrices are promising substrates for tissue generation by stem cells to replace poorly regenerating tissues such as cartilage. However, the dense matrix of decellularized cartilage impedes colonisation by stem cells. Here, we show that digestion of elastin fibre bundles traversing auricular cartilage creates channels through which cells can migrate into the matrix. Human chondrocytes and bone marrow-derived mesenchymal stromal cells efficiently colonise elastin-treated scaffolds through these channels, restoring a glycosaminoglycan-rich matrix and improving mechanical properties while maintaining size and shape of the restored tissue. The scaffolds are also rapidly colonised by endogenous cartilage-forming cells in a subcutaneously implanted osteochondral biopsy model. Creating channels for cells in tissue matrices may be a broadly applicable strategy for recellularization and restoration of tissue function.


Subject(s)
Ear Cartilage/cytology , Pancreatic Elastase/metabolism , Adolescent , Aged , Animals , Cattle , Child , Chondrogenesis , Elastin/metabolism , Extracellular Matrix/chemistry , Female , Glycosaminoglycans/metabolism , Humans , Mice, Nude , Middle Aged , Tissue Scaffolds/chemistry
4.
Eur Cell Mater ; 33: 43-58, 2017 01 30.
Article in English | MEDLINE | ID: mdl-28138954

ABSTRACT

Bone marrow derived mesenchymal stem cells (MSCs) have immunomodulatory and trophic capacities. For therapeutic application in local chronic inflammatory diseases, MSCs, preferably of allogeneic origin, have to retain immunomodulatory properties. This might be achieved by encapsulation of MSCs in a biomaterial that protects them from the host immune system. Most studies investigating the properties of MSCs for therapeutic application use short term cultures of cells in monolayer. Since the physical environment of MSCs can influence their functionality, we evaluated the feasibility of preserving the immunomodulatory properties of MSCs encapsulated in a three-dimensional alginate construct. After 5 weeks of implantation in immunocompetent rats, active allogeneic MSCs encapsulated in alginate were still detectable by Bio Luminescence Imaging and Magnetic Resonance Imaging of luciferase transduced and superparamagnetic iron oxide labelled MSCs. MSCs injected in saline were only detectable up to 1 week after injection. Moreover, the MSCs encapsulated in alginate responded to inflammatory stimuli similarly to MSCs in monolayer culture. In addition, MSC-alginate beads secreted immunomodulatory and trophic factors and inhibited T-cell proliferation after 30 d of in vitro culture. Our data indicate that allogeneic MSCs encapsulated in alginate persist locally and could act as an interactive immunomodulatory or trophic factor release system for several weeks, making this an interesting system to investigate for application in inflammatory disease conditions.


Subject(s)
Alginates/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Adipogenesis/drug effects , Animals , Cell Proliferation/drug effects , Cells, Immobilized/cytology , Cells, Immobilized/drug effects , Cells, Immobilized/metabolism , Culture Media, Conditioned/pharmacology , Gene Expression Regulation/drug effects , Glucuronic Acid/pharmacology , Hexuronic Acids/pharmacology , Humans , Immunocompetence/drug effects , Immunomodulation/drug effects , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Rats, Wistar , Subcutaneous Tissue/drug effects , Subcutaneous Tissue/pathology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Transplantation, Homologous
5.
Eur Cell Mater ; 27: 264-80; discussion 278-80, 2014 Apr 06.
Article in English | MEDLINE | ID: mdl-24706178

ABSTRACT

Cartilage has limited self-regenerative capacity. Tissue engineering can offer promising solutions for reconstruction of missing or damaged cartilage. A major challenge herein is to define an appropriate cell source that is capable of generating a stable and functional matrix. This study evaluated the performance of culture-expanded human chondrocytes from ear (EC), nose (NC) and articular joint (AC), as well as bone-marrow-derived and adipose-tissue-derived mesenchymal stem cells both in vitro and in vivo. All cells (≥ 3 donors per source) were culture-expanded, encapsulated in alginate and cultured for 5 weeks. Subsequently, constructs were implanted subcutaneously for 8 additional weeks. Before and after implantation, glycosaminoglycan (GAG) and collagen content were measured using biochemical assays. Mechanical properties were determined using stress-strain-indentation tests. Hypertrophic differentiation was evaluated with qRT-PCR and subsequent endochondral ossification with histology. ACs had higher chondrogenic potential in vitro than the other cell sources, as assessed by gene expression and GAG content (p < 0.001). However, after implantation, ACs did not further increase their matrix. In contrast, ECs and NCs continued producing matrix in vivo leading to higher GAG content (p < 0.001) and elastic modulus. For NC-constructs, matrix-deposition was associated with the elastic modulus (R² = 0.477, p = 0.039). Although all cells--except ACs--expressed markers for hypertrophic differentiation in vitro, there was no bone formed in vivo. Our work shows that cartilage formation and functionality depends on the cell source used. ACs possess the highest chondrogenic capacity in vitro, while ECs and NCs are most potent in vivo, making them attractive cell sources for cartilage repair.


Subject(s)
Alginates/pharmacology , Chondrogenesis , Hyaline Cartilage/cytology , Mesenchymal Stem Cell Transplantation , Regeneration , Adipose Tissue/cytology , Adolescent , Adult , Aged , Animals , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Collagen/metabolism , Glucuronic Acid/pharmacology , Glycosaminoglycans/metabolism , Hexuronic Acids/pharmacology , Humans , Hyaline Cartilage/metabolism , Hyaline Cartilage/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Middle Aged , Stress, Mechanical , Tissue Scaffolds/chemistry
6.
Eur Cell Mater ; 27: 112-23; discussion 123, 2014 Feb 03.
Article in English | MEDLINE | ID: mdl-24488855

ABSTRACT

Hydrogels pose interesting features for cartilage regeneration strategies, such as the option for injectability and in situ gelation resulting in optimal filling of defects. We aimed to study different hydrogels for their capability to support chondrogenesis of human bone marrow-derived mesenchymal stem cells (hBMSCs). hBMSCs were encapsulated in alginate, alginate with hyaluronic acid (alginate/HA), fibrin or thermoresponsive HA grafted with poly(N-isopropyl acrylamide) side-chains (HA-pNIPAM). Glycosaminoglycan production and cartilage-related gene expression were significantly higher in hBMSC-alginate and hBMSC-fibrin constructs than in the other constructs. Supplementation of alginate with HA was not beneficial. hBMSC-alginate, hBMSC-fibrin and hBMSC-HA-pNIPAM constructs were placed in simulated defects in osteochondral biopsies and cultured in vitro for 28 d. Biopsies containing hBMSC-alginate and hBMSC-fibrin were implanted subcutaneously in nude mice for 12 weeks. hBMSC-alginate constructs had significantly higher cartilage-related gene expression after 28 d of culture as well as significantly more safranin-O positive repair tissue after 12 weeks in vivo than hBMSC-fibrin constructs. Although initial experiments with hBMSC-hydrogel constructs suggested comparable results of hBMSC-alginate, hBMSC-fibrin and hBMSC-HA-pNIPAM constructs, culture in the osteochondral biopsy model in vitro as well as in vivo revealed differences, suggests that chondrogenesis of hBMSCs in an osteochondral environment is hydrogel-dependent.


Subject(s)
Chondrocytes/cytology , Chondrogenesis , Hydrogels/pharmacology , Mesenchymal Stem Cells/cytology , Acrylic Resins/pharmacology , Adult , Alginates/pharmacology , Animals , Cartilage/metabolism , Cartilage/physiology , Cattle , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Fibrin/pharmacology , Glucuronic Acid/pharmacology , Guided Tissue Regeneration , Hexuronic Acids/pharmacology , Humans , Hyaluronic Acid/pharmacology , Hydrogels/chemistry , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Osteochondrosis/surgery , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regeneration , Tissue Scaffolds/chemistry
7.
J Tissue Eng Regen Med ; 6(1): 68-76, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21305699

ABSTRACT

Human bone marrow stromal-derived mesenchymal stem cells (hBMSCs) will differentiate into chondrocytes in response to defined chondrogenic medium containing transforming growth factor-ß (TGFß). Results in the literature suggest that the three mammalian subtypes of TGFß (TGFß1, TGFß2 and TGFß3) provoke certain subtype-specific activities. Therefore, the aim of our study was to investigate whether the TGFß subtypes affect chondrogenic differentiation of in vitro cultured hBMSCs differently. HBMSC pellets were cultured for 5 weeks in chondrogenic media containing either 2.5, 10 or 25 ng/ml of TGFß1, TGFß2 or TGFß3. All TGFß subtypes showed a comparable dose-response curve, with significantly less cartilage when 2.5 ng/ml was used and no differences between 10 and 25 ng/ml. Four donors with variable chondrogenic capacity were used to evaluate the effect of 10 ng/ml of either TGFß subtype on cartilage formation. No significant TGFß subtype-dependent differences were observed in the total amount of collagen or glycosaminoglycans. Cells from a donor with low chondrogenic capacity performed equally badly with all TGFß subtypes, while a good donor overall performed well. After addition of ß-glycerophosphate during the last 2 weeks of culture, the expression of hypertrophy markers was analysed and mineralization was demonstrated by alkaline phosphatase activity and alizarin red staining. No significant TGFß subtype-dependent differences were observed in expression collagen type X or VEGF secretion. Nevertheless, pellets cultured with TGFß1 had significantly less mineralization than pellets cultured with TGFß3. In conclusion, this study suggests that TGFß subtypes do affect terminal differentiation of in vitro cultured hBMSCs differently.


Subject(s)
Bone Marrow Cells/cytology , Cartilage/metabolism , Mesenchymal Stem Cells/cytology , Stromal Cells/cytology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta/metabolism , Cell Culture Techniques/methods , Chondrocytes/cytology , Collagen Type X/metabolism , Dose-Response Relationship, Drug , Glycerophosphates/pharmacology , Humans , Tissue Engineering/methods , Vascular Endothelial Growth Factor A/metabolism
8.
Cartilage ; 1(3): 200-10, 2010 Jul.
Article in English | MEDLINE | ID: mdl-26069552

ABSTRACT

Cartilage oligomeric matrix protein (COMP) is a protein present in the cartilage matrix and is expressed more abundantly in osteoarthritis cartilage than in healthy cartilage. The present study was designed to investigate the effect of transforming growth factor ß (TGFß) on COMP deposition and the influence of COMP on collagen biochemistry in a long-term 3-dimensional culture. Bovine chondrocytes in alginate beads were cultured with or without 25 ng/mL TGFß2 for 21 or 35 days. COMP was overexpressed in bovine chondrocytes using lentiviral transfection. COMP gene expression, COMP protein production, collagen and proteoglycan deposition, and collagen fibril thickness were determined. Addition of TGFß2 resulted in more COMP mRNA and protein than the control condition without growth factors. Lentiviral transduction with COMP resulted in elevated gene expression of COMP and increased COMP levels in the alginate bead and culture medium compared to untransfected cells. Overexpression of COMP did not affect the deposition of collagen, collagen cross-linking, proteoglycan deposition, or the mechanical properties. Stimulating COMP production by either TGFß2 or lentivirus resulted in collagen fibrils with a smaller diameter. Taken together, COMP deposition can be modulated in cartilage matrix production by the addition of growth factors or by overexpression of COMP. Inducing COMP protein expression resulted in collagen fibrils with a smaller diameter. Because it has been demonstrated that the collagen fibril diameter is associated with mechanical functioning of the matrix, modulating COMP levels should be taken into account in cartilage regeneration strategies.

9.
J Tissue Eng Regen Med ; 3(2): 117-23, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19160351

ABSTRACT

The optimal stimulus to repair or regenerate cartilage is not known. We therefore modulated collagen deposition, collagen crosslinking and GAG deposition simultaneously during cartilage matrix production and integrative repair, creating more insight into their role in cartilage repair processes. Insulin-like growth factor 1 (IGF-1; increases proteoglycan and collagen synthesis), beta-aminopropionitrile (BAPN; a reversible inhibitor of collagen crosslinking) and para-nitrophenyl-beta-D-xyloside (PNPX; interferes with proteoglycan production) were used. Bovine articular chondrocytes were cultured in alginate beads for 3 weeks with or without IGF-1, BAPN or PNPX alone and in all possible combinations, followed by 3 weeks in control medium. DNA content, GAG and collagen deposition and collagen crosslinks were determined. Cartilage constructs were cultured under the same conditions and histologically analysed for integration of two opposing cartilage matrices. In alginate cultures, inhibition of collagen crosslinking with BAPN, in combination with promotion of matrix synthesis using IGF1, was most beneficial for matrix deposition. Addition of PNPX was always detrimental for matrix deposition. For integration of opposing cartilage constructs, the combination of BAPN, IGF1 and temporary prevention of proteoglycan formation with PNPX was most beneficial. When a new matrix is produced, proteoglycans are important to retain collagen in the matrix. When two already formed cartilage matrices have to integrate, a temporary absence of proteoglycans and temporary inhibition of collagen crosslinking might be more beneficial in combination with stimulation of collagen production, e.g. by IGF1. Therefore, the choice of soluble factors to promote cartilage regeneration depends on the type of therapy that will be used.


Subject(s)
Cartilage, Articular/growth & development , Proteoglycans/biosynthesis , Regeneration , Alginates , Animals , Cartilage, Articular/physiology , Cattle , Cells, Cultured , Glucuronic Acid , Hexuronic Acids
10.
Osteoarthritis Cartilage ; 16(3): 359-66, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17714957

ABSTRACT

BACKGROUND: Damage to articular cartilage is one of the features of osteoarthritis (OA). Cartilage damage is characterised by a net loss of collagen and proteoglycans. The collagen network is considered highly important for cartilage function but little is known about processes that control composition and function of the cartilage collagen network in cartilage tissue engineering. Therefore, our aim was to study the contribution of collagen amount and number of crosslinks on the functionality of newly formed matrix during cartilage repair. METHODS: Bovine articular chondrocytes were cultured in alginate beads. Collagen network formation was modulated using the crosslink inhibitor beta-aminopropionitrile (BAPN; 0.25mM). Constructs were cultured for 10 weeks with/without BAPN or for 5 weeks with BAPN followed by 5 weeks without. Collagen deposition, number of crosslinks and susceptibility to degradation by matrix metalloproteinase-1 (MMP-1) were examined. Mechanical properties of the constructs were determined by unconfined compression. RESULTS: BAPN for 5 weeks increased collagen deposition accompanied by increased construct stiffness, despite the absence of crosslinks. BAPN for 10 weeks further increased collagen amounts. Absence of collagen crosslinks did not affect stiffness but ability to hold water was lower and susceptibility to MMP-mediated degradation was increased. Removal of BAPN after 5 weeks increased collagen amounts, allowed crosslink formation and increased stiffness. DISCUSSION: This study demonstrates that both collagen amounts and its proper crosslinking are important for a functional cartilage matrix. Even in conditions with elevated collagen deposition, crosslinks are needed to provide matrix stiffness. Crosslinks also contribute to the ability to hold water and to the resistance against degradation by MMP-1.


Subject(s)
Aminopropionitrile/pharmacology , Chondrocytes , Extracellular Matrix/chemistry , Fibrillar Collagens/metabolism , Regeneration/physiology , Tissue Engineering/methods , Alginates , Animals , Biomechanical Phenomena , Cartilage, Articular/pathology , Cattle , Cell Count , Cells, Cultured , Extracellular Matrix/drug effects , Extracellular Matrix/enzymology , Gene Expression , Matrix Metalloproteinases/pharmacology , Osteoarthritis/pathology , Permeability , Phenotype , Stress, Mechanical , Time Factors
11.
Osteoarthritis Cartilage ; 14(11): 1136-46, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16730198

ABSTRACT

OBJECTIVE: Examine effects of insulin-like growth factor 1 (IGF1), transforming growth factor beta2 (TGFbeta2) and fibroblast growth factor 2 (FGF2) on proteoglycan and collagen network and biomechanical properties of the newly formed cartilage matrix. METHODS: Bovine articular chondrocytes were cultured in alginate beads for 3 weeks with or without FGF2, TGFbeta2 or IGF1 in the presence of 10% FCS. Proteoglycan content, collagen content, hydroxylysylpyridinoline cross-links and overall matrix metalloproteinase (MMP) activity in the culture medium were measured. Alginate disks cultured for 5 weeks were used to evaluate the effect of growth factors on mechanical properties of the construct by determining the equilibrium aggregate modulus and secant modulus. RESULTS: IGF1 increased collagen and proteoglycan deposition. FGF2 mainly decreased collagen deposition and TGFbeta2 proteoglycan deposition. A decrease in cross-links was observed in matrix produced by chondrocytes cultured in the presence of TGFbeta2. IGF1 and FGF2 had no influence on the number of cross-links per collagen molecule. Overall MMP activity was significantly higher in culture medium of cells cultured with FGF2. TGFbeta2 and IGF1 had no effect on MMP activity. After 35 days of culture, the matrix produced under influence of IGF1 had a lower permeability and a trend to increase stiffness. FGF2 showed a trend to lower both properties. TGFbeta2 had no effect on these parameters. CONCLUSION: IGF1, TGFbeta2 and FGF2 had differential effects on collagen network formation. Of the three growth factors tested, IGF1 seems to be best in promoting the formation of a functional collagen network since it increased proteoglycan and collagen deposition and improved the mechanical properties.


Subject(s)
Cartilage, Articular/metabolism , Collagen/analysis , Fibroblast Growth Factor 2/pharmacology , Insulin-Like Growth Factor I/pharmacology , Transforming Growth Factor beta2/pharmacology , Aggrecans/analysis , Animals , Biomechanical Phenomena , Cartilage, Articular/drug effects , Cattle , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Collagen Type II/analysis , DNA/analysis , Extracellular Matrix/drug effects , Extracellular Matrix/physiology , Forelimb , Gene Expression , Immunohistochemistry/methods , Matrix Metalloproteinases/metabolism , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/analysis , Protein-Lysine 6-Oxidase/analysis , Proteoglycans/analysis
12.
Leukemia ; 16(9): 1782-90, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12200694

ABSTRACT

The first definitive long-term repopulating hematopoietic stem cells (HSCs) emerge from and undergo rapid expansion in the embryonic aorta-gonad-mesonephros (AGM) region. To investigate the presumptive unique characteristics of the embryonic hematopoietic microenvironment and its surrounding tissues, we have generated stromal clones from subdissected day 10 and day 11 AGMs, embryonic livers (ELs) and gut mesentery. We here examine the ability of 19 of these clones to sustain extended long-term cultures (LTCs) of human CD34(+) umbilical cord blood (UCB) cells in vitro. The presence of in vitro repopulating cells was assessed by sustained production of progenitor cells (extended LTC-CFC) and cobblestone area-forming cells (CAFC). The embryonic stromal clones differed greatly in their support for human HSCs. Out of eight clones tested in the absence of exogenous cytokines, only one (EL-derived) clone was able to provide maintenance of HSCs. Addition of either Tpo or Flt3-L + Tpo improved the long-term support of about 50% of the tested clones. Cultures on four out of 19 clones, ie the EL-derived clone mentioned, two urogenital-ridge (UG)-derived clones and one gastrointestinal (GI)-derived clone, allowed a continuous expansion of primitive CAFC and CFU-GM with over several hundred-fold more CAFC(week6) produced in the 12th week of culture. This expansion was considerably higher than that found with the FBMD-1 cell line, which is appreciated by many investigators for its support of human HSCs, under comparable conditions. This stromal cell panel derived from the embryonic regions may be a powerful tool in dissecting the factors mediating stromal support for maintenance and expansion of HSCs.


Subject(s)
Embryo, Mammalian/cytology , Fetal Blood/cytology , Stem Cells/cytology , Stromal Cells/cytology , Animals , Antigens, CD34/metabolism , Aorta/embryology , Cell Lineage , Cell Separation , Clone Cells/cytology , Coculture Techniques , Colony-Forming Units Assay , Cytokines/metabolism , Digestive System/embryology , Gonads/embryology , Graft Survival , Hematopoiesis , Humans , Liver/embryology , Mesentery/embryology , Mesonephros/embryology , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/chemistry , Stem Cells/drug effects , Stromal Cells/metabolism
13.
Blood ; 96(2): 498-505, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10887111

ABSTRACT

Gap junctions (GJs) provide for a unique system of intercellular communication (IC) allowing rapid transport of small molecules from cell to cell. GJs are formed by a large family of proteins named connexins (Cxs). Cx43 has been considered as the predominantly expressed Cx by hematopoietic-supporting stroma. To investigate the role of the Cx family in hemopoiesis, we analyzed the expression of 11 different Cx species in different stromal cell lines derived from murine bone marrow (BM) or fetal liver (FL). We found that up to 5 Cxs are expressed in FL stromal cells (Cx43, Cx45, Cx30.3, Cx31, and Cx31.1), whereas only Cx43, Cx45, and Cx31 were clearly detectable in BM stromal cells. In vivo, the Cx43-deficient 14.5- to 15-day FL cobblestone area-forming cells (CAFC)-week 1-4 and colony-forming unit contents were 26%-38% and 39%-47% lower than in their wild-type counterparts, respectively. The reintroduction of the Cx43 gene into Cx43-deficient FL stromal cells was able to restore their diminished IC to the level of the wild-type FL stromal cells. In addition, these Cx43-reintroduced stromal cells showed an increased support ability (3.7-fold) for CAFC-week 1 in normal mouse BM and 5-fold higher supportive ability for CAFC-week 4 in 5-fluorouracil-treated BM cells as compared with Cx43-deficient FL stromal cells. These findings suggest that stromal Cx43-mediated IC, although not responsible for all GJ-mediated IC of stromal cells, plays a role in the supportive ability for hemopoietic progenitors and stem cells. (Blood. 2000;96:498-505)


Subject(s)
Connexin 43/physiology , Gap Junctions/physiology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/ultrastructure , Stromal Cells/metabolism , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/ultrastructure , Cell Line , Connexin 43/deficiency , Connexin 43/genetics , Fluoresceins/metabolism , Fluorescent Antibody Technique , Gene Expression , Hematopoiesis , Liver/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...