Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Mol Biol Rep ; 48(2): 1559-1565, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33398679

ABSTRACT

Mesenchymal stem cells (MSC) differentiate into different cell types and have immunomodulatory and paracrine effects. Cryopreservation of umbilical cord tissue as a source of MSC is very promising for regenerative medicine. We aim to evaluate a protocol for cryopreserving this tissue sectioned into small fragments with viable MSC. A total of 723 samples were frozen, thawed and cultured to obtain primary cultures of MSC. These were followed until 90-100% confluence and flow cytometric analysis were performed to confirm the mesenchymal phenotype. Samples in which protocol alterations at the collection of the samples were reported, were excluded for microbial contamination analysis leaving a total of 634 samples composed of 181 vaginal and 453 cesarean births. All cultures reach confluence with a media of 22.57 days and 97% in 28 or fewer days. Evaluated cultures showed low percentage of CD45+ and high of CD73 and CD90. Eight samples were subcultured 4 or 5 times and differentiated to chondrocytes and osteocytes to test differentiation potential with positive results. Umbilical cord tissue collections showed similar microbial profile and risk factors to those reported of umbilical cord blood collections, but with higher contamination frequencies. Cryopreserved tissue samples had viable cells that can be expanded without losing differentiation potential. Higher contamination frequencies compared to umbilical cord blood collection are not surprising, however, microbial load and survival of microorganisms to cryopreservation are expected to be lower.


Subject(s)
Cell Culture Techniques/methods , Cryopreservation/methods , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Cell Differentiation/genetics , Cell Proliferation/genetics , Chondrocytes/cytology , Fetal Blood/cytology , Humans , Osteocytes/cytology , Regenerative Medicine/methods
2.
Acta Histochem ; 122(8): 151636, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33132168

ABSTRACT

INTRODUCTION: Mesenchymal stem cells (MSCs) are characterized by the potential to differentiate into multiple cell lineages, high proliferation rates, and self-renewal capacity, in addition to the ability to maintain their undifferentiated state. These cells have been identified in physiological oral tissues such as pulp tissue, dental follicle, apical papilla and periodontal ligament, as well as in pathological situations such as chronic periapical lesions (CPLs). The criteria used for the identification of MSCs include the positive expression of specific surface antigens, with CD73, CD90, CD105, CD44, CD146, STRO-1, CD166, NANOG and OCT4 being the most specific for these cells. AIM: The aim of this review was to explore the literature on markers able to identify MSCs as well as the presence of these cells in the healthy periodontal ligament and CPLs, highlighting their role in regenerative medicine and implications in the progression of these lesions. METHODS: Narrative literature review searching the PubMed and Medline databases. Articles published in English between 1974 and 2020 were retrieved. CONCLUSION: The included studies confirmed the presence of MSCs in the healthy periodontal ligament and in CPLs. Several surface markers are used for the characterization of these cells which, although not specific, are effective in cell recognition. Mesenchymal stem cells participate in tissue repair, exerting anti- inflammatory, immunosuppressive and proangiogenic effects, and are therefore involved in the progression and attenuation of CPLs or even in the persistence of these lesions.


Subject(s)
Mesenchymal Stem Cells/cytology , Periapical Diseases/pathology , Periodontal Ligament/cytology , Regenerative Endodontics/methods , Adipocytes/cytology , Adipocytes/immunology , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Surface/genetics , Antigens, Surface/immunology , Biomarkers/metabolism , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , Chondrocytes/cytology , Chondrocytes/immunology , Dental Pulp/cytology , Dental Pulp/immunology , Gene Expression , Humans , Mesenchymal Stem Cells/immunology , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/immunology , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/immunology , Osteoblasts/cytology , Osteoblasts/immunology , Osteogenesis/genetics , Osteogenesis/immunology , Periapical Diseases/genetics , Periapical Diseases/immunology , Periapical Diseases/therapy , Periodontal Ligament/immunology
3.
Int J Dev Biol ; 64(7-8-9): 433-443, 2020.
Article in English | MEDLINE | ID: mdl-33063837

ABSTRACT

The neural crest (NC) is a transitory embryonic structure of vertebrates that gives rise to an astonishing variety of derivatives, encompassing both neural and mesenchymal cell types. Neural crest cells (NCCs) are an excellent model to study how environmental factors modulate features such as cell multipotentiality and differentiation. Tests with multifunctional substrates that allow NCCs to express their full potential, while promoting cell subcloning, are needed to advance knowledge about NCC self-renewal and to foster future biotechnological approaches. Here we show that a self-assembled peptide named PuraMatrixTM is an excellent substrate that allows the differentiation of NCCs based on the identification of seven different cell types. Depending on the PuraMatrixTM concentration employed, different frequencies and quantities of a given cell type were obtained. It is noteworthy that an enormous quantity and diversity of mesenchymal phenotypes, such as chondrocytes, could be observed. The quantity of adipocytes and osteocytes also increased with the use of mesenchymal differentiation factors (MDF), but PuraMatrixTM alone can support the appearance of these mesenchymal cell types. PuraMatrixTM will promote advances in studies related to multipotentiality, self-renewal and control of NCC differentiation, since it is an extremely simple and versatile material which can be employed for both in vivo and in vitro experiments.


Subject(s)
Cell Differentiation/physiology , Cell Self Renewal/physiology , Mesenchymal Stem Cells/physiology , Neural Crest/physiology , Peptides/metabolism , Adipocytes/cytology , Adipocytes/physiology , Animals , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/physiology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/physiology , Neural Crest/cytology , Osteocytes/cytology , Osteocytes/physiology , Quail/embryology , Quail/metabolism , Vertebrates/embryology , Vertebrates/metabolism
4.
Biomed Pharmacother ; 127: 110170, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32334373

ABSTRACT

BACKGROUND: Bushenhuoxue formula (BSHXF) has shown excellent clinical effects on the treatment of osteoporosis in China. The aim of this study is to determine the anti-osteoporosis effects and precise molecular mechanisms of BSHXF on mouse models. METHODS: Ten-week-old female C57BL/6 J mice were subjected to ovariectomy and provided a daily treatment of BSHXF. At 8 weeks post-surgery, the femurs were harvested for tissue analyses including µCT, histology, qRT-PCR and immunohistochemical (IHC) staining of ß-catenin, ALP and FABP4. To investigate the role of ß-catenin in the anti-osteoporosis effects of BSHXF, relative experiments mentioned above were performed in ß-catenin conditional knockout mice. RESULTS: Ovariectomized (OVX) mice presented severe bone loss and excessive fat accumulation in the chondro-osseous junction underneath the growth plate, with decreased expression of ALP and increased expression of FABP4. BSHXF significantly recovered the OVX-induced abnormal osteogenesis and adipogenesis with the activation of ß-catenin in growth plate chondrocytes. Further, we generated growth plate chondrocyte-specific ß-catenin knockout (ß-cateninGli1ER) mice that exhibited bone loss and fat accumulation in the chondro-osseous junction, similar to the OVX mice. However, BSHXF failed to rescue the osteoporosis-like phenotype in ß-cateninGli1ER mice, indicating the anti-osteoporosis effects of BSHXF act mainly through ß-catenin signaling. No significant restoration of ALP and FABP4 was observed in ß-cateninGli1ER mice after the treatment of BSHXF. CONCLUSIONS: BSHXF attenuates osteoporosis by promoting osteogenic differentiation of growth plate chondrocytes mainly in ß-catenin-dependent manner. BSHXF is considered as a new candidate for the treatment of osteoporosis.


Subject(s)
Chondrocytes/drug effects , Drugs, Chinese Herbal/pharmacology , Osteogenesis/drug effects , Osteoporosis/drug therapy , Adipogenesis/drug effects , Animals , Cell Differentiation/drug effects , Chondrocytes/cytology , Female , Growth Plate/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoporosis/pathology , Ovariectomy , Wnt Signaling Pathway/drug effects , beta Catenin/genetics , beta Catenin/metabolism
5.
Bioelectromagnetics ; 41(1): 41-51, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31736106

ABSTRACT

Magnetic fields (MFs) have been used as an external stimulus to increase cell proliferation in chondrocytes and extracellular matrix (ECM) synthesis of articular cartilage. However, previously published studies have not shown that MFs are homogeneous through cell culture systems. In addition, variables such as stimulation times and MF intensities have not been standardized to obtain the best cellular proliferative rate or an increase in molecular synthesis of ECM. In this work, a stimulation device, which produces homogeneous MFs to stimulate cell culture surfaces was designed and manufactured using a computational model. Furthermore, an in vitro culture of primary rat chondrocytes was established and stimulated with two MF schemes to measure both proliferation and ECM synthesis. The best proliferation rate was obtained with an MF of 2 mT applied for 3 h, every 6 h for 8 days. In addition, the increase in the synthesis of glycosaminoglycans was statistically significant when cells were stimulated with an MF of 2 mT applied for 5 h, every 6 h for 8 days. These findings suggest that a stimulation with MFs is a promising tool that could be used to improve in vitro treatments such as autologous chondrocyte implantation, either to increase cell proliferation or stimulate molecular synthesis. Bioelectromagnetics. 2020;41:41-51 © 2019 Bioelectromagnetics Society.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Extracellular Matrix/metabolism , Magnetic Fields/adverse effects , Animals , Cell Proliferation , Cell Survival , Cells, Cultured , Cells, Immobilized , Computer Simulation , Glycosaminoglycans/chemistry , Rats , Rats, Wistar , Surface Properties , Temperature , Time Factors
6.
Braz J Med Biol Res ; 52(9): e8525, 2019.
Article in English | MEDLINE | ID: mdl-31411316

ABSTRACT

Many compounds of ginsenosides show anti-inflammatory properties. However, their anti-inflammatory effects in intervertebral chondrocytes in the presence of inflammatory factors have never been shown. Increased levels of pro-inflammatory cytokines are generally associated with the degradation and death of chondrocytes; therefore, finding an effective and nontoxic substance that attenuates the inflammation is worthwhile. In this study, chondrocytes were isolated from the nucleus pulposus tissues, and the cells were treated with ginsenoside compounds and IL-1ß, alone and in combination. Cell viability and death rate were assessed by CCK-8 and flow cytometry methods, respectively. PCR, western blot, and immunoprecipitation assays were performed to determine the mRNA and protein expression, and the interactions between proteins, respectively. Monomeric component of ginsenoside Rd had no toxicity at the tested range of concentrations. Furthermore, Rd suppressed the inflammatory response of chondrocytes to interleukin (IL)-1ß by suppressing the increase in IL-1ß, tumor necrosis factor (TNF)-α, IL-6, COX-2, and inducible nitric oxide synthase (iNOS) expression, and retarding IL-1ß-induced degradation of chondrocytes by improving cell proliferation characteristics and expression of aggrecan and COL2A1. These protective effects of Rd were associated with ubiquitination of IL-1 receptor accessory protein (IL1RAP), blocking the stimulation of IL-1ß to NF-κB. Bioinformatics analysis showed that NEDD4, CBL, CBLB, CBLC, and ITCH most likely target IL1RAP. Rd increased intracellular ITCH level and the amount of ITCH attaching to IL1RAP. Thus, IL1RAP ubiquitination promoted by Rd is likely to occur by up-regulation of ITCH. In summary, Rd inhibited IL-1ß-induced inflammation and degradation of intervertebral disc chondrocytes by increasing IL1RAP ubiquitination.


Subject(s)
Chondrocytes/drug effects , Ginsenosides/pharmacology , Interleukin-1 Receptor Accessory Protein/metabolism , Interleukin-1beta/drug effects , Intervertebral Disc Degeneration/metabolism , Adult , Aged , Aggrecans/metabolism , Cell Survival/drug effects , Chondrocytes/cytology , Chondrocytes/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Female , Ginsenosides/metabolism , Humans , Inflammation/metabolism , Interleukin-1beta/metabolism , Low Back Pain/metabolism , Male , Middle Aged , Nitric Oxide Synthase/metabolism , Nucleus Pulposus/cytology , Nucleus Pulposus/drug effects , Nucleus Pulposus/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ubiquitination
7.
Tissue Eng Part B Rev ; 25(4): 357-373, 2019 08.
Article in English | MEDLINE | ID: mdl-30913997

ABSTRACT

Herein we review the state-of-the-art in tissue engineering for repair of articular cartilage. First, we describe the molecular, cellular, and histologic structure and function of endogenous cartilage, focusing on chondrocytes, collagens, extracellular matrix, and proteoglycans. We then explore in vitro cell culture on scaffolds, discussing the difficulties involved in maintaining or obtaining a chondrocytic phenotype. Next, we discuss the diverse compounds and designs used for these scaffolds, including natural and synthetic biomaterials and porous, fibrous, and multilayer architectures. We then report on the mechanical properties of different cell-loaded scaffolds, and the success of these scaffolds following in vivo implantation in small animals, in terms of generating tissue that structurally and functionally resembles native tissue. Last, we highlight future trends in this field. We conclude that despite major technical advances made over the past 15 years, and continually improving results in cartilage repair experiments in animals, the development of clinically useful implants for regeneration of articular cartilage remains a challenge


Subject(s)
Biocompatible Materials/chemistry , Cartilage, Articular/physiology , Chondrocytes/cytology , Regeneration , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cartilage, Articular/injuries , Extracellular Matrix , Humans , Wound Healing
8.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;52(9): e8525, 2019. tab, graf
Article in English | LILACS | ID: biblio-1011614

ABSTRACT

Many compounds of ginsenosides show anti-inflammatory properties. However, their anti-inflammatory effects in intervertebral chondrocytes in the presence of inflammatory factors have never been shown. Increased levels of pro-inflammatory cytokines are generally associated with the degradation and death of chondrocytes; therefore, finding an effective and nontoxic substance that attenuates the inflammation is worthwhile. In this study, chondrocytes were isolated from the nucleus pulposus tissues, and the cells were treated with ginsenoside compounds and IL-1β, alone and in combination. Cell viability and death rate were assessed by CCK-8 and flow cytometry methods, respectively. PCR, western blot, and immunoprecipitation assays were performed to determine the mRNA and protein expression, and the interactions between proteins, respectively. Monomeric component of ginsenoside Rd had no toxicity at the tested range of concentrations. Furthermore, Rd suppressed the inflammatory response of chondrocytes to interleukin (IL)-1β by suppressing the increase in IL-1β, tumor necrosis factor (TNF)-α, IL-6, COX-2, and inducible nitric oxide synthase (iNOS) expression, and retarding IL-1β-induced degradation of chondrocytes by improving cell proliferation characteristics and expression of aggrecan and COL2A1. These protective effects of Rd were associated with ubiquitination of IL-1 receptor accessory protein (IL1RAP), blocking the stimulation of IL-1β to NF-κB. Bioinformatics analysis showed that NEDD4, CBL, CBLB, CBLC, and ITCH most likely target IL1RAP. Rd increased intracellular ITCH level and the amount of ITCH attaching to IL1RAP. Thus, IL1RAP ubiquitination promoted by Rd is likely to occur by up-regulation of ITCH. In summary, Rd inhibited IL-1β-induced inflammation and degradation of intervertebral disc chondrocytes by increasing IL1RAP ubiquitination.


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Aged , Chondrocytes/drug effects , Ginsenosides/pharmacology , Interleukin-1beta/drug effects , Interleukin-1 Receptor Accessory Protein/metabolism , Intervertebral Disc Degeneration/metabolism , Dinoprostone/metabolism , Cell Survival/drug effects , Tumor Necrosis Factor-alpha/metabolism , Low Back Pain/metabolism , Nitric Oxide Synthase/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Ginsenosides/metabolism , Cyclooxygenase 2/metabolism , Aggrecans/metabolism , Interleukin-1beta/metabolism , Ubiquitination , Nucleus Pulposus/cytology , Nucleus Pulposus/drug effects , Nucleus Pulposus/metabolism , Inflammation/metabolism
9.
Biol Res ; 51(1): 41, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30384862

ABSTRACT

BACKGROUND: Osteoarthritis (OA) can be defined as degradation of articular cartilage of the joint, and is the most common degenerative disease. To regenerate the damaged cartilage, different experimental approaches including stem cell therapy have been tried. One of the major limitations of stem cell therapy is the poor post-transplantation survival of the stem cells. Anoikis, where insufficient matrix support and adhesion to extracellular matrix causes apoptotic cell death, is one of the main causes of the low post-transplantation survival rate of stem cells. Therefore, enhancing the initial interaction of the transplanted stem cells with chondrocytes could improve the therapeutic efficacy of stem cell therapy for OA. Previously, protein kinase C activator phorbol 12-myristate 13-acetate (PMA)-induced increase of mesenchymal stem cell adhesion via activation of focal adhesion kinase (FAK) has been reported. In the present study, we examine the effect PMA on the adipose-derived stem cells (ADSCs) adhesion and spreading to culture substrates, and further on the initial interaction between ADSC and chondrocytes. RESULTS: PMA treatment increased the initial adhesion of ADSC to culture substrate and cellular spreading with increased expression of adhesion molecules, such as FAK, vinculin, talin, and paxillin, at both RNA and protein level. Priming of ADSC with PMA increased the number of ADSCs attached to confluent layer of cultured chondrocytes compared to that of untreated ADSCs at early time point (4 h after seeding). CONCLUSION: Taken together, the results of this study suggest that priming ADSCs with PMA can increase the initial interaction with chondrocytes, and this proof of concept can be used to develop a non-invasive therapeutic approach for treating OA. It may also accelerate the regeneration process so that it can relieve the accompanied pain faster in OA patients. Further in vivo studies examining the therapeutic effect of PMA pretreatment of ADSCs for articular cartilage damage are required.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/cytology , Protein Kinase C/pharmacology , Stem Cells/drug effects , Blotting, Western , Cell Adhesion , Cell Communication , Cell Culture Techniques , Cell Differentiation , Cell Survival , Chondrocytes/drug effects , Humans , Reverse Transcriptase Polymerase Chain Reaction
10.
Stem Cell Rev Rep ; 14(5): 734-743, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29728886

ABSTRACT

PURPOSE: Chondral lesion is a pathology with high prevalence, reaching as much as 63% of general population and 36% among athletes. The ability of human Dental Pulp Stem Cells (DPSCs) to differentiate into chondroblasts in vitro suggests that this stem cell type may be useful for tissue bioengineering. However, we have yet to identify a study of large animal models in which DPSCs were used to repair articular cartilage. Therefore, this study aimed to describe a novel treatment for cartilage lesion with DPSCs on a large animal model. METHODS: Mesenchymal stem cells (MSC) were obtained from deciduous teeth and characterized by flow cytometry. DPSCs were cultured and added to a collagen type I/III biomaterial composite scaffold. Brazilian miniature pig (BR-1) was used. A 6-mm diameter, full-thickness chondral defect was created in each posterior medial condyle. The defects were covered with scaffold alone or scaffold + DPSCs on the contralateral side. Animals were euthanized 6 weeks post-surgery. Cartilage defects were analyzed macroscopically and histology according to modified O'Driscoll scoring system. RESULTS: Flow cytometry confirmed characterization of DPSCs as MSCs. Macroscopic and histological findings suggested that this time period was reasonable for evaluating cartilage repair. To our knowledge, this study provides the first description of an animal model using DPSCs to study the differentiation of hyaline articular cartilage in vivo. CONCLUSION: The animals tolerated the procedure well and did not show clinical or histological rejection of the DPSCs, reinforcing the feasibility of this descriptive miniature pig model for pre-clinical studies.


Subject(s)
Cartilage Diseases/therapy , Cartilage, Articular/growth & development , Mesenchymal Stem Cell Transplantation , Stem Cells/cytology , Animals , Cartilage Diseases/physiopathology , Cartilage, Articular/cytology , Cell Differentiation/genetics , Chondrocytes/cytology , Chondrogenesis/genetics , Dental Pulp/cytology , Humans , Mesenchymal Stem Cells/cytology , Swine , Swine, Miniature , Tissue Engineering , Tooth, Deciduous/cytology
11.
Clinics (Sao Paulo) ; 73: e268, 2018.
Article in English | MEDLINE | ID: mdl-29641802

ABSTRACT

OBJECTIVES: Articular cartilage is vulnerable to injuries and undergoes an irreversible degenerative process. The use of amniotic fluid mesenchymal stromal stem cells for the reconstruction of articular cartilage is a promising therapeutic alternative. The aim of this study was to investigate the chondrogenic potential of amniotic fluid mesenchymal stromal stem cells from human amniotic fluid from second trimester pregnant women in a micromass system (high-density cell culture) with TGF-ß3 for 21 days. METHODS: Micromass was performed using amniotic fluid mesenchymal stromal stem cells previously cultured in a monolayer. Chondrocytes from adult human normal cartilage were used as controls. After 21 days, chondrogenic potential was determined by measuring the expression of genes, such as SOX-9, type II collagen and aggrecan, in newly differentiated cells by real-time PCR (qRT-PCR). The production of type II collagen protein was observed by western blotting. Immunohistochemistry analysis was also performed to detect collagen type II and aggrecan. This study was approved by the local ethics committee. RESULTS: SOX-9, aggrecan and type II collagen were expressed in newly differentiated chondrocytes. The expression of SOX-9 was significantly higher in newly differentiated chondrocytes than in adult cartilage. Collagen type II protein was also detected. CONCLUSION: We demonstrate that stem cells from human amniotic fluid are a suitable source for chondrogenesis when cultured in a micromass system. amniotic fluid mesenchymal stromal stem cells are an extremely viable source for clinical applications, and our results suggest the possibility of using human amniotic fluid as a source of mesenchymal stem cells.


Subject(s)
Cell Culture Techniques/methods , Chondrocytes/cytology , Chondrogenesis , Mesenchymal Stem Cells/cytology , Aggrecans/metabolism , Amniotic Fluid , Cell Differentiation , Collagen Type II/analysis , Female , Gene Expression , Humans , Pregnancy , SOX9 Transcription Factor/metabolism , Transforming Growth Factor beta3/metabolism
12.
J Pharm Pharmacol ; 70(1): 89-100, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29105086

ABSTRACT

OBJECTIVES: Verify the in-vitro effect of triiodothyronine (T3) on the chondrogenic differentiation of female rat bone marrow mesenchymal stem cells (BMMSCs) over several time periods and at several doses. METHODS: CD54 + /CD73 + /CD90 +  BMMSCs from Wistar female rats were cultured in chondrogenic medium with or without T3 (0.01; 1; 100; 1000 nm). At seven, 14 and 21 days, the cell morphology, chondrogenic matrix formation and expression of Sox9 and collagen II were evaluated. KEY FINDINGS: The dose of 100 nm did not alter the parameters evaluated in any of the periods studied. However, the 0.01 nm T3 dose improved the chondrogenic potential by increasing the chondrogenic matrix formation and expression of Sox9 and collagen II in at least one of the evaluated periods; the 1 nm T3 dose also improved the chondrogenic potential by increasing the chondrogenic matrix formation and the expression of collagen II in at least one of the evaluated periods. The 1000 nm T3 dose improved the chondrogenic potential by increasing the chondrogenic matrix formation and Sox9 expression in at least one of the evaluated periods. CONCLUSIONS: T3 has a dose-dependent effect on the differentiation of BMMSCs from female rats.


Subject(s)
Cell Differentiation/drug effects , Chondrogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Triiodothyronine/pharmacology , Animals , Cells, Cultured , Chondrocytes/cytology , Collagen Type II/genetics , Dose-Response Relationship, Drug , Female , Mesenchymal Stem Cells/cytology , Rats , Rats, Wistar , SOX9 Transcription Factor/genetics , Time Factors , Triiodothyronine/administration & dosage
13.
Biol. Res ; 51: 41, 2018. graf
Article in English | LILACS | ID: biblio-983943

ABSTRACT

BACKGROUND: Osteoarthritis (OA) can be defined as degradation of articular cartilage of the joint, and is the most common degenerative disease. To regenerate the damaged cartilage, different experimental approaches including stem cell therapy have been tried. One of the major limitations of stem cell therapy is the poor post-transplantation survival of the stem cells. Anoikis, where insufficient matrix support and adhesion to extracellular matrix causes apoptotic cell death, is one of the main causes of the low post-transplantation survival rate of stem cells. Therefore, enhancing the initial interaction of the transplanted stem cells with chondrocytes could improve the therapeutic efficacy of stem cell therapy for OA. Previously, protein kinase C activator phorbol 12-myristate 13-acetate (PMA)- induced increase of mesenchymal stem cell adhesion via activation of focal adhesion kinase (FAK) has been reported. In the present study, we examine the effect PMA on the adipose-derived stem cells (ADSCs) adhesion and spreading to culture substrates, and further on the initial interaction between ADSC and chondrocytes. RESULTS: PMA treatment increased the initial adhesion of ADSC to culture substrate and cellular spreading with increased expression of adhesion molecules, such as FAK, vinculin, talin, and paxillin, at both RNA and protein level. Priming of ADSC with PMA increased the number of ADSCs attached to confluent layer of cultured chondrocytes compared to that of untreated ADSCs at early time point (4 h after seeding). CONCLUSION: Taken together, the results of this study suggest that priming ADSCs with PMA can increase the initial interaction with chondrocytes, and this proof of concept can be used to develop a non-invasive therapeutic approach for treating OA. It may also accelerate the regeneration process so that it can relieve the accompanied pain faster in OA patients. Further in vivo studies examining the therapeutic effect of PMA pretreatment of ADSCs for articular cartilage damage are required.


Subject(s)
Humans , Stem Cells/drug effects , Protein Kinase C/pharmacology , Cartilage, Articular/cytology , Chondrocytes/cytology , Cell Adhesion , Cell Communication , Cell Differentiation , Cell Survival , Blotting, Western , Cell Culture Techniques , Chondrocytes/drug effects , Reverse Transcriptase Polymerase Chain Reaction
14.
Clinics ; Clinics;73: e268, 2018. tab, graf
Article in English | LILACS | ID: biblio-890754

ABSTRACT

OBJECTIVES: Articular cartilage is vulnerable to injuries and undergoes an irreversible degenerative process. The use of amniotic fluid mesenchymal stromal stem cells for the reconstruction of articular cartilage is a promising therapeutic alternative. The aim of this study was to investigate the chondrogenic potential of amniotic fluid mesenchymal stromal stem cells from human amniotic fluid from second trimester pregnant women in a micromass system (high-density cell culture) with TGF-β3 for 21 days. METHODS: Micromass was performed using amniotic fluid mesenchymal stromal stem cells previously cultured in a monolayer. Chondrocytes from adult human normal cartilage were used as controls. After 21 days, chondrogenic potential was determined by measuring the expression of genes, such as SOX-9, type II collagen and aggrecan, in newly differentiated cells by real-time PCR (qRT-PCR). The production of type II collagen protein was observed by western blotting. Immunohistochemistry analysis was also performed to detect collagen type II and aggrecan. This study was approved by the local ethics committee. RESULTS: SOX-9, aggrecan and type II collagen were expressed in newly differentiated chondrocytes. The expression of SOX-9 was significantly higher in newly differentiated chondrocytes than in adult cartilage. Collagen type II protein was also detected. CONCLUSION: We demonstrate that stem cells from human amniotic fluid are a suitable source for chondrogenesis when cultured in a micromass system. amniotic fluid mesenchymal stromal stem cells are an extremely viable source for clinical applications, and our results suggest the possibility of using human amniotic fluid as a source of mesenchymal stem cells.


Subject(s)
Humans , Pregnancy , Cell Culture Techniques/methods , Chondrocytes/cytology , Chondrogenesis , Mesenchymal Stem Cells/cytology , Gene Expression , Cell Differentiation , Collagen Type II/analysis , Aggrecans/metabolism , Transforming Growth Factor beta3/metabolism , SOX9 Transcription Factor/metabolism , Amniotic Fluid
15.
Cell Tissue Bank ; 18(3): 369-381, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28584920

ABSTRACT

To compare the quality of the repair tissue in three-dimensional co-culture of human chondrocytes implanted in an in vivo model. Six cadaveric and five live human donors were included. Osteochondral biopsies from the donor knees were harvested for chondrocyte isolation. Fifty percent of cadaveric chondrocytes were expanded until passage-2 (P2) while the remaining cells were cryopreserved in passage-0 (P0). Fresh primary chondrocytes (P0f) obtained from live human donors were co-cultured. Three-dimensional constructs were prepared with a monolayer of passage-2 chondrocytes, collagen membrane (Geistlich Bio-Gide®), and pellet of non-co-cultured (P2) or co-cultured chondrocytes (P2 + P0c, P2 + P0f). Constructs were implanted in the subcutaneous tissue of athymic mice and left for 3 months growth. Safranin-O and Alcian blue staining were used to glycosaminoglycan content assessment. Aggrecan and type-II collagen were evaluated by immunohistochemistry. New-formed tissue quality was evaluated with an adaptation of the modified O'Driscoll score. Histological quality of non-co-cultured group was 4.37 (SD ±4.71), while co-cultured groups had a mean score of 8.71 (SD ±3.98) for the fresh primary chondrocytes and 9.57 (SD ±1.27) in the cryopreserved chondrocytes. In immunohistochemistry, Co-culture groups were strongly stained for type-II and aggrecan not seen in the non-co-cultured group. It is possible to isolate viable chondrocytes from cadaveric human donors in samples processed in the first 48-h of dead. There is non-significant difference between the numbers of chondrocytes isolated from live or cadaveric donors. Cryopreservation of cadaveric primary chondrocytes does not alter the capability to form cartilage like tissue. Co-culture of primary and passaged chondrocytes enhances the histological quality of new-formed tissue compared to non-co-cultured cells.


Subject(s)
Cell Dedifferentiation , Chondrocytes/cytology , Chondrocytes/transplantation , Coculture Techniques/methods , Animals , Cadaver , Cartilage/cytology , Cells, Cultured , Glycosaminoglycans/analysis , Humans , Living Donors , Male , Mice, Nude , Tissue Engineering/methods , Wound Healing
16.
Int J Biol Macromol ; 103: 1019-1031, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28536017

ABSTRACT

The aim of the present study was to investigate the activities of natural chondroitin sulfates (CS) with different structures on cultured chondrocytes and macrophages. CS were isolated from cartilages of bovine trachea (BT), porcine trachea (PT), chicken sternum (Ch) and skate (Sk). The preparations were 90-98% pure, with ∼1% proteins, nucleic acids and keratan sulfate contaminants. Structural analysis of these CS and of commercial chondroitin 4- and 6-sulfate (C4S, C6S) have shown that most of their disaccharides are monosulfated, with varying proportions of 4- and 6-sulfation, and 2-7% non-sulfated disaccharides. Sk-CS and C6S contained detectable amounts of disulfated disaccharides. All the CS were polydisperse, with modal molecular weights of 26-135kDa. These CS had anti-inflammatory activities on both chondrocytes and macrophages, but with different efficiencies. On horse and human chondrocytes, they reduced the IL-1ß-induced liberation of NO and PGE2, and on RAW 264.7 immortalized macrophage-like cell line, C4S, C6S, Ch and Sk-CS decreased the LPS-induced liberation of TNF-α, but did not affect IL-6. In contrast, on bone marrow derived macrophages, C4S, C6S, BT and PT-CS reduced the LPS-induced liberation of TNF-α, IL-6, IL-1ß and NO, indicating that the RAW response to CS was different from that of primary macrophages.


Subject(s)
Chondrocytes/drug effects , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Macrophages/drug effects , Animals , Bone Marrow Cells/cytology , Chondrocytes/cytology , Interleukin-1beta/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Mice , RAW 264.7 Cells , Structure-Activity Relationship
17.
Tissue Eng Part A ; 23(17-18): 935-945, 2017 09.
Article in English | MEDLINE | ID: mdl-28384053

ABSTRACT

Injectable hydrogels have gained prominence in the field of tissue engineering for minimally invasive delivery of cells for tissue repair and in the filling of irregular defects. However, many injectable hydrogels exhibit long gelation times or are not stable for long periods after injection. To address these concerns, we used thermosensitive poly(N-vinylcaprolactam) (PNVCL) hydrogels due to their cytocompatibility and fast response to temperature stimuli. Changes in the PNVCL molecular weight and concentration enabled the development of hydrogels with tunable mechanical properties and fast gelation times (<60 s when the temperature was raised from room temperature to physiologic temperature). Chondrocytes (CHs) and mesenchymal stem cells were encapsulated in PNVCL hydrogels and exhibited high viability (∼90%), as monitored by Live/Dead staining and Alamar Blue assays. Three-dimensional constructs of CH-laden PNVCL hydrogels supported cartilage-specific extracellular matrix production both in vitro and after subcutaneous injection in nude rats for up to 8 weeks. Moreover, biochemical analyses of constructs demonstrated a time-dependent increase in glycosaminoglycans (GAGs) and collagen, which were significantly augmented in the implants cultured in vivo. Histological analyses also demonstrated regular distribution of synthesized cartilage components, including abundant GAGs and type II collagen. The findings from this study demonstrate thermosensitive PNVCL as a candidate injectable biomaterial to deliver cells for cartilage tissue engineering.


Subject(s)
Caprolactam/analogs & derivatives , Cartilage/metabolism , Chondrocytes/metabolism , Hydrogels/chemistry , Polymers/chemistry , Tissue Engineering/methods , Animals , Caprolactam/chemistry , Caprolactam/pharmacology , Cartilage/cytology , Cattle , Chondrocytes/cytology , Chondrocytes/transplantation , Hydrogels/pharmacology , Polymers/pharmacology , Rats , Rats, Nude
18.
Aging Male ; 20(3): 161-167, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28332902

ABSTRACT

This study aimed to analyze the effects of caloric restriction on aged femoral articular cartilage of Wistar rats. Three groups of eight animals each were considered: young (YC) and old (OC) control groups fed with a normal diet and old caloric restriction group (OCR) composed of 18-month-old animals fed with a 31% less caloric diet from 6-months of age. Articular cartilage was studied through morphometry and immunohistochemistry. Body mass was 12% less in the OCR group than in the OC group. The articular cartilage from OC rats show thinner medial condyles, fewer chondrocytes, smaller chondrocytes nuclear volume and, in both condyles, a predominance of collagen type II and less collagen density compared to both YC and OCR groups (p < .001). In contrast, OCR articular cartilage show thicker medial condyles, larger chondrocytes nuclear volume and increased collagen density compared to OC group (p < 0.001). We concluded that caloric restriction minimizes the effects of aging on medial condyles of the femoral articular cartilage.


Subject(s)
Aging/metabolism , Caloric Restriction , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Femur/pathology , Aging/physiology , Animals , Cartilage, Articular/cytology , Cartilage, Articular/pathology , Chondrocytes/cytology , Chondrocytes/pathology , Collagen/metabolism , Collagen/physiology , Male , Random Allocation , Rats , Rats, Wistar
19.
Stem Cell Res Ther ; 8(1): 27, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28173846

ABSTRACT

BACKGROUND: Stem cells are capable of unlimited self-renewal and are able to remain undifferentiated for extended periods of time prior to their differentiation into specific cell lineages. Because of the issues (ethical and religious) involved in the use of embryonic stem cells and the limited plasticity of adult stem cells, an alternative cell source could be foetal stem cells derived from extra-embryonic tissue, which are highly proliferative, grow in vitro and possess interesting immunogenic characteristics. As a result, the amniotic membrane of several species has been studied as an important new source of stem cells. METHODS: Here, we cultured and characterized mesenchymal progenitor cells derived from the rabbit amniotic membrane, and investigated their differentiation potential. In total, amniotic membranes were collected from eight rabbit foetuses and were isolated by the explant technique. The obtained cells were cultured in DMEM-HIGH glucose and incubated at 37 °C in a humidified atmosphere with 5% CO2. RESULTS: The cells adhered to the culture plates and showed a high proliferative capacity with fibroblast-like morphologies. The cells showed a positive response for markers for the cytoskeleton, mesenchymal stem cells and proliferation, pluripotency and haematopoietic precursor stem cells. However, the cells were negative for CD45, a marker of haematopoietic cells. Furthermore, the cells had the capacity to be induced to differentiate into osteogenic, adipogenic and chondrogenic lineages. In addition, when the cells were injected into nude mice, we did not observe the formation of tumours. CONCLUSIONS: In summary, our results demonstrate that multipotent mesenchymal stem cells can be obtained from the rabbit amniotic membrane for possible use in future cell therapy applications.


Subject(s)
Adipocytes/cytology , Amnion/cytology , Chondrocytes/cytology , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Osteoblasts/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Amnion/drug effects , Amnion/metabolism , Animals , Cell Differentiation , Cell Proliferation , Chondrocytes/drug effects , Chondrocytes/metabolism , Culture Media/pharmacology , Glucose/metabolism , Glucose/pharmacology , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Phenotype , Primary Cell Culture , Rabbits
20.
Dev Genes Evol ; 227(2): 121-127, 2017 03.
Article in English | MEDLINE | ID: mdl-27909803

ABSTRACT

The vertebrate endoskeleton results from the piecemeal assembly of bone and cartilage as well as additional types of calcified extracellular matrices produced by seemingly hybrid cell types of intermediate phenotypes between osteoblasts and chondrocytes. Hence, shedding light on the emergence and subsequent diversification of skeletal tissues represents a major challenge in vertebrate evolutionary developmental biology. A 150-year-old debate in the field was recently solved by lineage tracing experiments demonstrating that, during mouse endochondral bone development, a subset of chondrocytes evades apoptosis and transdifferentiates into osteoblasts at the chondro-osseous junction. Here, we interpret these new data from a broad phylogenetic perspective, integrating fossil, histological, cellular, and genetic evidence from a wide range of vertebrates. We propose a testable scenario according to which transdifferentiation played a fundamental role in the emergence of endochondral ossification, an osteichthyan-specific evolutionary novelty. The remarkable skeletal cell plasticity might be contingent on the similar architectures of the osteoblastic and chondrocytic gene regulatory networks, thereby providing a unifying mechanism underlying both complete transdifferentiation as well as partial cell type conversions observed in intermediate skeletal tissues such as the chondroid bone or globuli ossei.


Subject(s)
Cell Transdifferentiation , Evolution, Molecular , Osteogenesis/genetics , Animals , Chondrocytes/cytology , Osteoblasts/cytology
SELECTION OF CITATIONS
SEARCH DETAIL