Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Knee Surg Sports Traumatol Arthrosc ; 23(5): 1384-92, 2015 May.
Article in English | MEDLINE | ID: mdl-24221246

ABSTRACT

PURPOSE: Differences in the tissue-specific collagen maturation process between tendon and ligament are still unknown. Collagen cross-link formation is crucial for the collagen maturation process. The aim of this study is to examine collagen maturation processes of anterior cruciate ligament (ACL), medial collateral ligament (MCL), and patellar tendon (PT) in vitro, in order to determine the optimal cell source for tissue engineering of ligament. METHODS: Cells derived from the ACL, MCL, and PT of New Zealand white rabbits were isolated. Each cell type was cultured for up to 4 weeks after reaching confluence. Cell-matrix layers were evaluated and compared for their morphology, collagen cross-links, and gene expression levels of lysine hydroxylase 1 and 2, lysyl oxidase (LOX), tenomodulin, collagen1A1 (Col1A1), and collagen3A1 (Col3A1). RESULTS: Transmission electron microscopy photomicrographs verified that collagen fibrils were secreted from all three types of fibroblasts. A higher ratio of dihydroxylysinonorleucine/hydroxylysinonorleucine was evident in the ligament compared to the tendon, which was consistent with lysine hydroxylase 2/lysine hydroxylase 1 gene expression. The gene expression of LOX, which regulates the total amount of enzymatic cross-linking, and the gene expression levels of Col1A1 and Col3A1 were higher in the ACL matrix than in the MCL and PT matrices. CONCLUSION: ACL, MCL, and PT cells have distinct collagen maturation processes at the cellular level. In addition, the collagen maturation of ACL cells is not necessarily inferior to that of MCL and PT cells in that all three cell types have a good ability to synthesize collagen and induce collagen maturation. This bioactivity of ACL cells in terms of ligament-specific mature collagen induction can be applied to tissue-engineered ACL reconstruction or remnant preserving procedure with ACL reconstruction.


Subject(s)
Anterior Cruciate Ligament/cytology , Collagen/genetics , Extracellular Matrix/genetics , Fibroblasts/metabolism , Gene Expression Regulation , Medial Collateral Ligament, Knee/metabolism , Tendons/metabolism , Animals , Anterior Cruciate Ligament/metabolism , Cells, Cultured , Collagen/biosynthesis , Collateral Ligaments , DNA/genetics , Extracellular Matrix/metabolism , Female , Fibroblasts/ultrastructure , Medial Collateral Ligament, Knee/cytology , Microscopy, Electron, Transmission , Rabbits , Real-Time Polymerase Chain Reaction , Tendons/cytology , Tissue Engineering/methods
2.
Mol Cell Biochem ; 360(1-2): 235-42, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21938405

ABSTRACT

The adult human anterior cruciate ligament (ACL) has a poor functional healing response, whereas the medial collateral ligament (MCL) does not. The difference in intrinsic properties of these ligament cells can be due to their different response to their located microenvironment. Hypoxia is a key environmental regulator after ligament injury. In this study, we investigated the differential response of ACL and MCL fibroblasts to hypoxia on hypoxia-inducible factor-1α, vascular endothelial growth factor, and matrix metalloproteinase-2 (MMP-2) expression. Our results show that ACL cells responded to hypoxia by up-regulating the HIF-1α expression significantly as compared to MCL cells. We also observed that in MCL fibroblasts response to hypoxia resulted in increase in expression of VEGF as compared to ACL fibroblasts. After hypoxia treatment, mRNA and protein levels of MMP-2 increased in both ACL and MCL. Furthermore we found in ACL pro-MMP-2 was converted more into active form. However, hypoxia decreased the percentage of wound closure for both ligament cells and had a greater effect on ACL fibroblasts. These results demonstrate that ACL and MCL fibroblasts respond differently under the hypoxic conditions suggesting that these differences in intrinsic properties may contribute to their different healing responses and abilities.


Subject(s)
Anterior Cruciate Ligament/cytology , Gene Expression , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Matrix Metalloproteinase 2/metabolism , Medial Collateral Ligament, Knee/cytology , Vascular Endothelial Growth Factor A/metabolism , Adult , Cell Hypoxia , Cell Movement , Cells, Cultured , Cobalt , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibroblasts/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Matrix Metalloproteinase 2/genetics , Middle Aged , Vascular Endothelial Growth Factor A/genetics , Wound Healing , Young Adult
3.
Int Orthop ; 36(1): 207-13, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21674292

ABSTRACT

PURPOSE: The lysyl oxidase (LOX) family plays a crucial role in the formation and stabilisation of extracellular matrix (ECM) by catalysing the cross-linking of collagen and elastin, implicating its important fundamental roles in injury healing. A high level of transforming growth factor-ß(1) (TGF-ß(1)) accompanies the inflammatory phase of an injury of the knee joint. Our purpose was to detect the expressions of the LOX family in anterior cruciate ligament (ACL) and medial collateral ligament (MCL) response to TGF-ß(1). METHODS: This study used reversed transcript PCR, real time quantitative PCR and Western blot for analyses. RESULTS: The results showed significant increases in mRNA levels of LOX family members. At 5 ng/ml concentration of TGF-ß(1,) the gene profiles of LOXs showed most active, and LOX and LOXL-3 showed increasing peaks at 12 hours after TGF-ß(1) treatment (LOX: 7.2, 8.8-fold and LOXL-3: 3.8, 5.4-fold compared with normal controls in ACL and MCL, respectively); LOXL-1, LOXL-2 and LOXL-4 reached their highest amounts at six hours (LOXL-1: 1.9, 2.4-fold; LOXL-2: 14.8, 16.2-fold; LOXL-4: 2.5, 4.4-fold in ACL and MCL, respectively). Protein assays revealed that LOXs in ACL cells had relatively lower response to TGF-ß(1) compared with those in MCL cells. CONCLUSIONS: The differential expression and activities of LOXs might help to explain the intrinsic difference between ACL and MCL, and LOXs could imply a potential capability for ACL healing.


Subject(s)
Anterior Cruciate Ligament/cytology , Fibroblasts/cytology , Medial Collateral Ligament, Knee/cytology , Protein-Lysine 6-Oxidase/genetics , Transforming Growth Factor beta/pharmacology , Up-Regulation/drug effects , Adult , Animals , Blotting, Western , Cells, Cultured , Dose-Response Relationship, Drug , Female , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression Regulation/drug effects , Humans , Male , Mice , Middle Aged , Polymerase Chain Reaction , Protein-Lysine 6-Oxidase/metabolism
4.
Biochem Biophys Res Commun ; 391(1): 1142-7, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20006576

ABSTRACT

Anterior cruciate ligament (ACL)-derived cells have a character different from medial collateral ligament (MCL)-derived cells. However, the critical difference between ACL and MCL is still unclear in their healing potential and cellular response. The objective of this study was to investigate the mesenchymal differentiation property of each ligament-derived cell. Both ligament-derived cells differentiated into adipogenic, osteogenic, and chondrogenic lineages. In chondrogenesis, ACL-derived cells had the higher chondrogenic property than MCL-derived cells. The chondrogenic marker genes, Sox9 and alpha1(II) collagen (Col2a1), were induced faster in ACL-derived pellets than in MCL-derived pellets. Sox9 expression preceded the increase of Col2a1 in both pellet-cultured cells. However, the expression level of Sox9 and a ligament/tendon transcription factor Scleraxis did not parallel the increase of Col2a1 expression along with chondrogenic induction. The present study demonstrates that the balance between Sox9 and Scleraxis have an important role in the chondrogenic differentiation of ligament-derived cells.


Subject(s)
Anterior Cruciate Ligament/cytology , Cell Differentiation , Cell Lineage , Chondrogenesis , Animals , Anterior Cruciate Ligament/metabolism , Biomarkers/metabolism , Cells, Cultured , Collagen Type II/metabolism , Medial Collateral Ligament, Knee/cytology , Medial Collateral Ligament, Knee/metabolism , Mesoderm/cytology , Rabbits , SOX9 Transcription Factor/metabolism
5.
Biochem Biophys Res Commun ; 402(2): 329-34, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20937261

ABSTRACT

Basic fibroblast growth factor (bFGF) and growth and differentiation factor (GDF)-5 stimulate the healing of medial collateral ligament (MCL) injury. However, the effect of isolated and combined use of bFGF/GDF-5 remains still unclear. We investigated cellular proliferation and migration responding to bFGF/GDF-5 using rabbit MCL fibroblasts. Rabbit MCL injury was treated by bFGF and/or GDF-5 with peptide hydrogels. Gene expression and deposition of collagens in healing tissues were evaluated. bFGF/GDF-5 treatment additively enhanced cell proliferation and migration. bFGF/GDF-5 hydrogels stimulated Col1a1 expression without increasing Col3a1 expression. Combined use of bFGF/GDF-5 stimulated type I collagen deposition and the reorganization of fiber alignment, and induced better morphology of fibroblasts in healing MCLs. Our study indicates that combined use of bFGF/GDF-5 might enhance MCL healing by increasing proliferation and migration of MCL fibroblasts, and by regulating collagen synthesis and connective fiber alignment.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Growth Differentiation Factor 5/pharmacology , Medial Collateral Ligament, Knee/drug effects , Medial Collateral Ligament, Knee/injuries , Wound Healing/drug effects , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen Type I/metabolism , Female , Fibroblasts/drug effects , Medial Collateral Ligament, Knee/cytology , Rabbits
6.
Food Funct ; 11(3): 2654-2666, 2020 Mar 26.
Article in English | MEDLINE | ID: mdl-32159191

ABSTRACT

Osteoarthritis (OA), a progressive joint disorder, is principally characterized by the degeneration and destruction of articular cartilage. Previous research studies demonstrated that inflammation and ECM degradation play a major role in OA development. Hesperetin, the aglycone of neohesperidin found in the peel of Citrus aurantium L. (Rutaceae), demonstrated in several studies potential anti-inflammatory activity in a variety of diseases. However, the mechanisms by which hesperetin plays a protective role in osteoarthritis (OA) are not completely understood. In this study, we found the anti-inflammatory effects of hesperetin in the progression of OA in both in vitro and in vivo experiments. In vitro, IL-1ß-induced expression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), nitric oxide (NO), tumor necrosis factor alpha (TNF-α), prostaglandin E2 (PGE2), and interleukin-6 (IL-6) were inhibited by hesperetin. Moreover, hesperetin down-regulated the IL-1ß-stimulated matrix metalloproteinase-13 (MMP-13) and thrombospondin motifs 5 (ADAMTS-5) while up-regulating collagen type II and aggrecan. Mechanistically, we revealed that hesperetin suppressed nuclear factor kappa B (NF-κB) signaling by activating the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in IL-1ß-induced chondrocytes. Hesperetin-induced repression of OA development is shown using a DMM model. Taken together, our findings suggest that hesperetin may be a novel potential therapeutic agent for repressing the development of OA.


Subject(s)
Hesperidin/pharmacology , Osteoarthritis/metabolism , Protective Agents/pharmacology , Aged , Animals , Cells, Cultured , Chondrocytes/drug effects , Female , Humans , Interleukin-1beta/metabolism , Male , Medial Collateral Ligament, Knee/cytology , Mice , Mice, Inbred C57BL , Middle Aged , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects
7.
Connect Tissue Res ; 50(1): 7-13, 2009.
Article in English | MEDLINE | ID: mdl-19212848

ABSTRACT

The human ACL (anterior cruciate ligament) is susceptible to injury but has poor healing response, whereas an injured MCL (medial collateral ligament) can be repaired relatively well. Since MMPs (matrix metalloproteases) and TIMPs (tissue inhibitor of metalloproteases) are involved in this tissue remodeling process, investigation of different response of MMPs and TIMPs family in ACL and MCL fibroblasts might lead to understanding the differential matrix remodeling process as well as their different healing ability. The first step would be determination of whether these tissue remodeling effectors are present in ligaments. In this study, we designed primers for real-time RT-PCR and determined the expression of MMPs and TIMPs family in ACL and MCL fibroblasts with synovium as a positive control. Semiquantitative RT-PCR revealed that multiple MMPs and TIMPs expressed in human ACL and MCL fibroblasts except MMP-8, 10, 12, 13, 15, 16, 20, and 26. MMP-7 was present in MCL but not in ACL fibroblast. Quantitative real-time RT-PCR showed that mRNA levels of MMP-1, 2, 14, 17, 23A, and 23B and TIMP-4 are significantly higher in MCL than in ACL fibroblasts. However, MMP-3 is higher in ACL than in MCL fibroblasts. We conclude that numerous MMPs and TIMPs family members that are differentially expressed in ACL and MCL might be involved in the differential matrix remodeling process as well as the differential healing ability of ACL and MCL.


Subject(s)
Anterior Cruciate Ligament/cytology , Fibroblasts/metabolism , Matrix Metalloproteinases/metabolism , Medial Collateral Ligament, Knee/cytology , RNA, Messenger/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Wound Healing/physiology , Cells, Cultured , DNA Primers/genetics , Humans , Reverse Transcriptase Polymerase Chain Reaction
8.
Undersea Hyperb Med ; 34(3): 181-90, 2007.
Article in English | MEDLINE | ID: mdl-17672174

ABSTRACT

PURPOSE: This study investigated hyperbaric oxygen (HBO2) and platelet-derived growth factor-BB (PDGF-BB) to determine their combined effects on fibroblasts from rabbit medial collateral ligament (MCL). METHOD: Cells were divided into four groups: (I) Control, (II) HBO2 treatment, (III) PDGF-BB treatment and (IV) HBO2 combined with PDGF-BB treatment. All hyperoxic cells were exposed to 100% O2 at 2.5 atmospheres absolute (ATA) in a hyperbaric chamber for 120 minutes per 48 hours. Measurement of cell growth was based on increase in cell number. Cell cycle modulations were analyzed by fluorescence-activated cell sorter (FACS). Quantity of Type I and Type III collagen was determined by western blotting and image analyzer. RESULTS: Treatment doses of HBO2 alone or PDGF-bb alone dependently increased cell growth. A combination of HBO2 treatment plus PDGF-bb treatment had an additive effect on cell growth in comparison with HBO2 treatment alone or PDGF-bb treatment alone. FACS analysis revealed that HBO2 alone, PDGF-bb alone and PDGF-bb plus HBO2 treatment increase the percentage of cells accumulated in S-phase. Western blotting analysis revealed that Type III collagen content was decreased significantly after HBO2 treatment alone or HBO2 plus PDGF-bb treatment but not in PDGF-bb treatment alone. In contrast, although Type I collagen content was increased after HBO2 treatment, the increase in Type I collagen (increase /original) was not statistically significant. CONCLUSION: HBO2 or HBO2 plus PDGF-bb treatment decreases the Type III collagen/Type I collagen content, which could result in mechanically stronger collagen fibrils. We propose HBO2 therapy as a potentially effective treatment for MCL healing.


Subject(s)
Fibroblasts/drug effects , Hyperbaric Oxygenation , Medial Collateral Ligament, Knee/cytology , Oxygen/pharmacology , Platelet-Derived Growth Factor/pharmacology , Animals , Becaplermin , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/metabolism , Collagen Type III/metabolism , Fibroblasts/cytology , Proto-Oncogene Proteins c-sis , Rabbits
9.
Cell Commun Adhes ; 13(3): 139-49, 2006.
Article in English | MEDLINE | ID: mdl-16798614

ABSTRACT

The interconnectivity of fibroblasts within the ligamentous extracellular matrix has been largely overlooked. Studies on the cell-to-cell contacts with their neighbors via gap junctions in ligament fibroblasts, and works on the ability of fibroblasts to generate interconnected networks in vivo, suggest interfibroblastic interactions play an important role in fundamental biological processes, including homeostasis and wound healing. The current study examines how fluidic shear stresses imposed by internal flow can be used to mediate the formation of three-dimensional, interconnected fibroblast networks within collagen solutions. Several fibroblast-collagen solutions were exposed to shear stresses via Poiselle Flow. The consequent changes in cell networking, interconnections, and cell morphology within collagen matrixes exhibited by cells derived from Bovine Medial Collateral Ligaments were analyzed. Results illustrate that higher imposed stresses generate cells with more dendritic and/or branched morphologies, which form more visible three-dimensional networks within collagen matrixes than fibroblast-collagen solutions that were unexposed to shear stress.


Subject(s)
Cell Surface Extensions/metabolism , Collagen/metabolism , Fibroblasts/cytology , Medial Collateral Ligament, Knee/cytology , Microfluidics , Animals , Cattle , Cells, Cultured , Gels , Solutions , Stress, Mechanical
10.
J Orthop Res ; 24(7): 1521-34, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16705720

ABSTRACT

The reason that the anterior cruciate ligament (ACL) has a very poor healing potential after injury is not well understood. In this study, we investigated the role of nitric oxide (NO) in the apoptotic cell death of ACL cells using a rabbit model and in vitro cell culture. The apoptosis of ACL cells in vivo was analyzed by TUNEL assay and electron microscopy. NO synthase (NOS) expression was observed by immunohistochemical analysis. ACL cells were cultured and the susceptibility to NO-induced apoptosis was tested. Inducible NOS (iNOS) expression after treatment with cytokines was examined by immunohistochemical and RT-PCR analyses. Mitogen-activated protein kinase (MAPK) inhibitors were used for the analysis of downstream signals. A significant number of apoptotic cells were observed on days 1 to 3 after injury; the apoptotic rate returned to the control level by day 7. Upregulation of iNOS in the ACL remnant was observed at day 1. Intraarticular injection of NOS inhibitor suppressed the apoptotic rate. Isolated ACL cells showed much higher susceptibility to NO-induced apoptosis than did medial collateral ligament cells. IL-1beta stimulated ACL cells to upregulate iNOS mRNA and increase NO production. p38 MAPK inhibitor decreased NO-induced apoptosis. Rapid iNOS induction after injury contributes to the high apoptotic rate of ACL cells, and this may partly account for the poor healing capacity of this ligament. iNOS and NO production is suggested to be stimulated by IL-1beta, and NO activates the p38 MAPK pathway and triggers an apoptotic signal in ACL cells.


Subject(s)
Anterior Cruciate Ligament/physiopathology , Apoptosis/physiology , Knee Injuries/physiopathology , Nitric Oxide Synthase Type II/metabolism , Animals , Anterior Cruciate Ligament/cytology , Anterior Cruciate Ligament/metabolism , Anterior Cruciate Ligament Injuries , Disease Models, Animal , Fibroblasts/physiology , Humans , Interleukin-1/physiology , Knee Injuries/metabolism , Medial Collateral Ligament, Knee/cytology , Medial Collateral Ligament, Knee/physiology , Nitric Oxide/physiology , Rabbits , Up-Regulation , p38 Mitogen-Activated Protein Kinases/physiology
11.
J Orthop Res ; 24(2): 149-58, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16435352

ABSTRACT

The role of cell surface integrins in cell migration, proliferation, and attachment to matrix molecules is well known. Integrin-matrix interactions have been implicated in mechanotransduction and load transmission from the outside to the inside of the cell. In this study, the effect of cyclic strain on the cell proliferation, attachment, and expression of integrin subunits beta1, beta3, and alpha5 was determined in anterior cruciate ligament (ACL) and medial collateral ligament (MCL) fibroblasts grown on polystyrene, Type I collagen, laminin, elastin, and fibronectin. ACL fibroblast proliferation was not affected by growth substrate whereas MCL cells reached confluence more rapidly on fibronectin compared with collagen or polystyrene. Exposure to 5% cyclic strain resulted in a significant decrease in ACL and MCL fibroblast proliferation on fibronectin and Type I collagen. MCL cells showed a greater strain-dependent inhibition of cells grown on a fibronectin substrate than those grown on collagen. This matrix-dependent effect of strain on cell proliferation was not seen with ACL cells. Attachment of ACL and MCL fibroblasts was stronger to fibronectin compared with Type I collagen, laminin, and polystyrene. In the absence of applied load, the expression of beta1, beta3, and alpha5 subunits was not substrate dependent and the expression of beta1 and alpha5 integrin subunits was higher in MCL cells than ACL cells on all substrates. In contrast, the expression of beta3 integrin subunit was higher in ACL cells than MCL cells. In response to 5% strain, beta1, and alpha5 expression increased in all fibroblasts with MCL cells having a higher magnitude of expression. beta3 expression showed a 90% increase in response to load when grown on laminin for both MCL and ACL fibroblasts and demonstrated no change in expression on Type I collagen or fibronectin. The duration of applied strain from 2 versus 22 h had no effect on cell proliferation or integrin expression.


Subject(s)
Anterior Cruciate Ligament/cytology , Fibroblasts/cytology , Integrins/metabolism , Mechanotransduction, Cellular/physiology , Medial Collateral Ligament, Knee/cytology , Stifle , Animals , Anterior Cruciate Ligament/drug effects , Anterior Cruciate Ligament/metabolism , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Dogs , Extracellular Matrix Proteins/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Mechanotransduction, Cellular/drug effects , Medial Collateral Ligament, Knee/drug effects , Medial Collateral Ligament, Knee/metabolism , Stress, Mechanical
12.
J Biomech ; 39(2): 276-83, 2006.
Article in English | MEDLINE | ID: mdl-16321629

ABSTRACT

This study quantified the apparent and intrinsic hydraulic permeability of human medial collateral ligament (MCL) under direct permeation transverse to the collagen fiber direction. A custom permeation device was built to apply flow across cylindrical samples of ligament while monitoring the resulting pressure gradient. MCLs from 5 unpaired human knees were used (donor age 55 +/- 16 yr, 4 males, 1 female). Permeability measurements were performed at 3 levels of compressive pre-strain (10%, 20% and 30%) and 5 pressures (0.17, 0.34, 1.03, 1.72 and 2.76 MPa). Apparent permeability was determined from Darcy's law, while intrinsic permeability was determined from the zero-pressure crossing of the pressure-permeability curves at each compressive pre-strain. Resulting data were fit to a finite deformation constitutive law [Journal of Biomechanics 23 (1990) 1145-1156]. The apparent permeability of human MCL ranged from 0.40 +/- 0.05 to 8.60 +/- 0.77 x 10(-16) m(4)/Ns depending on pre-strain and pressure gradient. There was a significant decrease in apparent permeability with increasing compressive pre-strain (p=0.024) and pressure gradient (p<0.001), and there was a significant interaction between the effects of compressive pre-strain and pressure (p<0.001). Intrinsic permeability was 14.14 +/- 0.74, 6.30 +/- 2.13 and 4.29 +/- 1.71 x 10(-16) m(4)/Ns for compressive pre-strains of 10%, 20% and 30%, respectively. The intrinsic permeability showed a faster decrease with increasing compressive pre-strain than that of bovine articular cartilage. These data provide a baseline for investigating the effects of disease and chemical modification on the permeability of ligament and the data should also be useful for modeling the poroelastic material behavior of ligaments.


Subject(s)
Body Water/physiology , Fibrillar Collagens/physiology , Fibrillar Collagens/ultrastructure , Medial Collateral Ligament, Knee/cytology , Medial Collateral Ligament, Knee/physiology , Weight-Bearing/physiology , Anisotropy , Cadaver , Compressive Strength/physiology , Elasticity , Female , Humans , In Vitro Techniques , Male , Middle Aged , Permeability , Stress, Mechanical
13.
Cell Signal ; 9(8): 587-94, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9429762

ABSTRACT

This study examines the real-time intracellular calcium concentration, [Ca2+]i, response of canine medial collateral ligament (MCL) and anterior cruciate ligament (ACL) fibroblasts subjected to a fluid-induced shear stress of 25 dynes/cm2. In experiments using a modified Hanks' Balanced Salt Solution (HBSS) perfusate, both cell types demonstrated a significant increase in peak [Ca2+]i compared to respective no-flow controls, the response of MCL fibroblasts being nearly 2-fold greater than that of ACL fibroblasts. In studies where the cells were bathed in a medium of HBSS supplemented with 2% newborn bovine serum (NBS) and then introduced to flow with the same medium, ACL fibroblasts responded nearly 3-fold greater than MCL fibroblasts. Neomycin (10 mM), thapsigarigin (1 microM) and Ca(2+)-free media supplemented with EGTA (1 mM) were able to inhibit significantly the [Ca2+]i response to flow with HBSS in both fibroblasts. Thapsigargin also blocked the NBS flow response in both cell types, while neomycin and Ca(2+)-free media significantly inhibited the ACL response. Our findings demonstrate that ACL and MCL cells are not the same. These differences may be related to the disparate healing capacity of the ACL and MCL observed clinically.


Subject(s)
Anterior Cruciate Ligament/cytology , Calcium/metabolism , Fibroblasts/metabolism , Medial Collateral Ligament, Knee/cytology , Animals , Buffers , Cattle , Cells, Cultured , Dogs , Egtazic Acid/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Intracellular Fluid , Stress, Physiological
14.
J Orthop Res ; 23(4): 949-57, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16023011

ABSTRACT

Recent studies have revealed that following injuries, ligament tissues such as anterior cruciate ligaments (ACL), release large amounts of matrix metalloproteinases (MMPs). These enzymes have a devastating effect on the healing process of the injured ligaments. Although these enzymes are produced following ligament injuries, because of different healing capacities seen between the medial collateral ligament (MCL) and ACL, we were curious to find if the MMP activity was expressed and modulated differently in these tissues. For this purpose ACL and MCL fibroblasts were seeded on equi-biaxial stretch chambers and were stretched in different levels. The stretched cells were assayed using Zymography, Western Blot and global MMP activity assays. The results showed that within 72 h after injurious stretch, production of 72 kD pro-MMP-2 increased in both ACL and MCL. However, the ACL fibroblasts generated significantly more pro-MMP-2 than the MCL fibroblasts. Furthermore we found in ACL pro-MMP-2 was converted more into active form. With 4-aminophenyl mercuric acetate (APMA) treatment, large amounts of pro-MMP-2 were converted into active form in both. This indicates that there is no significant difference between ACL and MCL fibroblasts in post-translational modification of MMP-2. The fluorescent MMP activity assays revealed that the MMP family activities were higher in the injured ACL fibroblasts than the MCL. Since the MMPs are critically involved in extracellular matrix (ECM) turnover, these findings may explain one of the reasons why the injured ACL hardly repairs. The higher levels of active MMP-2 seen in the ACL injuries may disrupt the delicate balance of ECM remodeling process. These results suggest that the generation and modulation of MMP-2 may be directly involved in the different responses seen in ACL and MCL injuries.


Subject(s)
Anterior Cruciate Ligament Injuries , Anterior Cruciate Ligament/enzymology , Matrix Metalloproteinase 2/metabolism , Medial Collateral Ligament, Knee/enzymology , Medial Collateral Ligament, Knee/injuries , Adult , Anterior Cruciate Ligament/cytology , Cells, Cultured , Culture Media, Conditioned/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Humans , In Vitro Techniques , Medial Collateral Ligament, Knee/cytology , Middle Aged , Phenylmercuric Acetate/analogs & derivatives , Phenylmercuric Acetate/pharmacology , Stress, Mechanical , Sulfhydryl Reagents/pharmacology
15.
Mech Ageing Dev ; 97(2): 121-30, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9226631

ABSTRACT

In this study, we examined the effects of age on collagen and total protein synthesis by ligament fibroblasts in response to growth factors. Three different doses of transforming growth factor-beta 1 (TGF-beta 1) or epidermal growth factor (EGF) were individually added to in vitro fibroblast cultures from the medial collateral ligament (MCL) of skeletally immature (age 3 months), mature (age 12 months) and senescent (age 48-51 months) rabbits. Analysis of the effects of age revealed that fibroblasts from senescent rabbits produced significantly less collagen in response to TGF-beta 1 or EGF stimulation when compared to fibroblasts from immature rabbits. Furthermore, increased age was found to result in significant reductions in the baseline levels of collagen synthesis but not total protein synthesis. Additionally, collagen and total protein synthesis by MCL fibroblasts were significantly affected by the TFG-beta 1 dose, but not by the EGF dose. When fibroblasts were normalized to their own controls, the increase in collagen and total protein synthesis due to TGF-beta 1 and EGF for the senescent group were found to be greater than those for the skeletally immature rabbits at all doses. This demonstrates that MCL fibroblasts from senescent rabbits are responsive to growth factors.


Subject(s)
Aging/metabolism , Collagen/biosynthesis , Epidermal Growth Factor/pharmacology , Protein Biosynthesis , Transforming Growth Factor beta/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Medial Collateral Ligament, Knee/cytology , Rabbits
16.
Tissue Eng ; 5(5): 443-52, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10586100

ABSTRACT

Tissue engineering offers the possibility of replacing damaged human ligaments with engineered ligament tissues. Hence, we attempted to culture in vitro ligament tissues by seeding human anterior cruciate ligament (ACL) and medial collateral ligament (MCL) cells onto synthetic biodegradable polymer fiber scaffolds. The ACL and MCL cells readily attached to the scaffold fibers. These cells and their secreted matrix soon surrounded the scaffold fibers and bridged the gaps in between. Beginning at 2 weeks, portions of the scaffolds were completely filled with tissue matrix. By 5 weeks, the scaffolds became single bundles of tissue. Thus the cell/fiber system appears to be a viable system for culturing ligament tissues. Additionally, cell proliferation under mechanical and biochemical stimuli was studied for up to 4 days. Whereas mechanical stimulus and transforming growth factor enhanced proliferation, inflammatory agents (lipopolysaccharide and complement C5a) had a negative effect. This work can thus contribute to a sound strategy for culturing replacement ligament tissues in vitro.


Subject(s)
Anterior Cruciate Ligament/cytology , Artificial Organs , Cell Culture Techniques/methods , Medial Collateral Ligament, Knee/cytology , Anterior Cruciate Ligament/drug effects , Anterior Cruciate Ligament Injuries , Biodegradation, Environmental , Complement C5a/pharmacology , Humans , Lipopolysaccharides/pharmacology , Medial Collateral Ligament, Knee/drug effects , Medial Collateral Ligament, Knee/injuries , Stress, Mechanical , Transforming Growth Factor beta/pharmacology
17.
Microsc Res Tech ; 58(1): 39-44, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12112421

ABSTRACT

A potential strategy to enhance ligament healing by transplantation of mesenchymal stem cells (MSCs), which are demonstrated to differentiate into fibroblast-like cells in vitro, is presented. The objective of this study was to follow transplanted nucleated cells from bone marrow, which contain MSCs, in the healing medial collateral ligament (MCL) over time, and to examine their phenotype and survivability. It was hypothesized that MSCs in nucleated cells from bone marrow would differentiate into fibroblast-like cells in the healing ligament following adaptation to the environment. The transplantation model employed in this study eliminates the immune response to a donor by the recipient using a transgenic rat (donor), which does not produce foreign protein from transgenes, and its wild-type rat (recipient) in order to simulate autologous transplantation. The MCL of the wild-type rat was ruptured, where 1 x 10(6) nucleated cells of bone marrow from the transgenic rat were injected. The transgenes in transplanted nucleated cells were detected throughout the healing MCL for 28 days by in situ hybridization. At 3 days, many donor cells were evident in the injury site and fascial pocket, and some were found in the midsubstance. Morphologically, transplanted cells with elongated nuclei were found at the ruptured edge of the midsubstance and surface of the unruptured site after 3 days. At 28 days, these cells continued to survive in the healing MCL. Their shapes were similar to those of surrounding recipient MCL fibroblasts. Thus, transplanted cells might differentiate into fibroblasts. Therefore, it was demonstrated that there is a potential for nucleated cells from bone marrow to serve as a vehicle for therapeutic molecules as well as to be a source in enhancing healing of ligaments.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Transplantation , Medial Collateral Ligament, Knee/cytology , Medial Collateral Ligament, Knee/surgery , Animals , Animals, Genetically Modified , Cell Count , Female , Fibroblasts/cytology , Genetic Therapy , Genetic Vectors , Medial Collateral Ligament, Knee/injuries , Mesoderm/cytology , Models, Animal , Rats , Transplantation, Autologous , Wound Healing
18.
J Orthop Res ; 13(2): 166-73, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7722753

ABSTRACT

The adhesiveness of fibroblasts from the human anterior cruciate and medial collateral ligaments to the laminin molecule was studied, with particular emphasis on the intrinsic differences between fibroblasts from the two ligaments. Cellular adhesion strength, adhesion area, laminin concentration, and seeding time were examined. Cell adhesion to laminin anchored with poly-D-lysine to a cleaned cover glass was measured with a micropipette micromanipulation system after seeding. The adhesion strength of fibroblasts from the anterior cruciate ligament to laminin was greater than and significantly different from that of fibroblasts from the medial collateral ligament, depending on the laminin concentration. Fibroblasts from the anterior cruciate ligament also exhibited an increase in adhesion strength, dependent on laminin concentration of as much as 30 micrograms/ml, at which the laminin receptors were thought to be saturated. Fibroblasts from the medial collateral ligament did not show such an increase except at laminin concentrations of 5-10 micrograms/ml. There was no significant difference in adhesion area between fibroblasts from the two ligaments except after 45 minutes at a laminin concentration of 40 micrograms/ml. For both, the adhesion to laminin showed little correlation to seeding time during periods of as long as 60 minutes. Measurements of adhesion area also failed to show a significant correlation to seeding time for fibroblasts from either ligament at laminin concentrations of 20 and 40 micrograms/ml. Adhesion strength normalized by adhesion area had no correlation to seeding time.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Anterior Cruciate Ligament/cytology , Fibroblasts/physiology , Laminin/physiology , Medial Collateral Ligament, Knee/cytology , Anterior Cruciate Ligament/physiology , Cell Adhesion/physiology , Humans , Medial Collateral Ligament, Knee/physiology , Micromanipulation
19.
J Orthop Res ; 13(3): 435-41, 1995 May.
Article in English | MEDLINE | ID: mdl-7602405

ABSTRACT

Cellular migration and proliferation are integral aspects of wound healing. An in vitro assay for cellular migration and proliferation using explants of rabbit anterior cruciate and medial collateral ligaments was developed previously. This study presents the effects of serum-free culture medium supplemented with basic fibroblast growth factor, bovine insulin, transforming growth factor-beta 1, and platelet-derived growth factor-B, added either individually or in combination, on cell outgrowth in explants of rabbit anterior cruciate and medial collateral ligaments. Outgrowth was assessed at 3 and 6 days by counting the number of cells surrounding the tissue explants. For explants of both ligaments, cell outgrowth was dependent on the presence of 10% fetal bovine serum or the combination of growth factors. Little outgrowth occurred in explants of either ligament in the presence of basic fibroblast growth factor, transforming growth factor-beta 1, or bovine insulin. Platelet-derived growth factor-B at concentrations of 20 and 100 ng/ml seemed to increase cell outgrowth from medial collateral ligament explants at 6 days. The outgrowth from the explants of both ligaments was much greater in the presence of all four growth factors than the sum of the outgrowth with the individual factors. This synergistic effect was as much as 10-fold and 20-fold for the anterior cruciate ligament explants at days 3 and 6, respectively, but no more than 3-fold for the medial collateral ligament explants at these times. Medial collateral ligament explants exhibited greater cell outgrowth than anterior cruciate ligament explants in 10% serum and in the presence of the four growth factors.


Subject(s)
Anterior Cruciate Ligament/cytology , Growth Substances/pharmacology , Medial Collateral Ligament, Knee/cytology , Animals , Cell Count/drug effects , Cell Division/drug effects , Culture Techniques , Drug Synergism , Male , Rabbits , Time Factors
20.
J Orthop Res ; 13(2): 174-83, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7722754

ABSTRACT

The differential capacities of the anterior cruciate and medial collateral ligaments to heal may be related to differences in cellular function. This study tested the hypothesis that differential expression of integrins occurs in these ligaments after injury. The integrins are a family of cell surface receptors that mediate adhesion, migration, and other cellular functions critical to the healing of a wound. A similar complement and amount of the beta 1 subfamily of integrins are known to be present on the unperturbed anterior cruciate and medial collateral ligaments in humans and rabbits. A partial laceration was surgically created in these two ligaments in 12 anesthetized New Zealand White rabbits. Immunohistochemistry was performed on sections from the ligaments at 1, 3, 7, and 10 days after injury, using monoclonal antibodies directed against the integrin subunits beta 1, alpha 5, alpha 6, and alpha v. Between 3 and 7 days, the wounded medial collateral ligament demonstrated a striking increase in staining for the beta 1, alpha 5, and alpha v subunits on the fibroblasts, within the repair site, and on capillary endothelium. Increased staining was most marked for the beta 1 subunit and less marked for the alpha 5 and alpha v subunits. The alpha 6 subunit stained exclusively vascular structures within the healing medial collateral ligament. In marked contrast, the anterior cruciate ligament, which does not mount an effective repair response, demonstrated no comparable alteration of integrin expression from baseline levels.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Anterior Cruciate Ligament Injuries , Integrins/metabolism , Medial Collateral Ligament, Knee/injuries , Wound Healing/physiology , Animals , Anterior Cruciate Ligament/cytology , Anterior Cruciate Ligament/metabolism , Antibodies, Monoclonal , Fibroblasts/metabolism , Immunohistochemistry , Inflammation/metabolism , Inflammation/physiopathology , Medial Collateral Ligament, Knee/cytology , Medial Collateral Ligament, Knee/metabolism , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL