Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Sci Food Agric ; 103(14): 6884-6894, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37286475

ABSTRACT

BACKGROUND: Porcine nasal cartilage type II collagen-derived peptides (PNCPs) may be complexed with calcium to provide a highly bioavailable, low-cost, and effective calcium food supplement. However, the calcium-binding characteristics of PNCPs have not yet been investigated. In the present study, calcium-binding peptides were derived from porcine nasal cartilage type II collagen and the resulting PNCPs-Ca complex was characterized. RESULTS: The study reveals that the calcium-binding capacity of PNCPs is closely related to enzymatic hydrolysis conditions. The highest calcium-binding capacity of PNCPs was observed at a hydrolysis time of 4 h, temperature of 40 °C, enzyme dosage of 1%, and solid-to-liquid ratio of 1:10. Scanning electron microscopy and energy dispersive X-ray spectroscopy revealed that the PNCPs had a pronounced capacity for calcium binding, with the PNCPs-Ca complex exhibiting a clustered structure consisting of aggregated spherical particles. Fourier-transform infrared spectroscopy, fluorescence spectroscopy, X-ray diffraction, dynamic light scattering, amino acid composition, and molecular weight distribution analyses all indicated that the PNCPs and calcium complexed via the carboxyl oxygen and amino nitrogen atoms, leading to the formation of a ß-sheet structure during the chelation process. In addition, the stability of the PNCPs-Ca complex was maintained over a range of pH values consistent with those found in the human gastrointestinal tract, facilitating calcium absorption. CONCLUSION: These research findings suggest the feasibility of converting by-products from livestock processing into calcium-binding peptides, providing a scientific basis for the development of novel calcium supplements and the potential reduction of resource waste. © 2023 Society of Chemical Industry.


Subject(s)
Calcium , Nasal Cartilages , Humans , Animals , Swine , Calcium/metabolism , Collagen Type II , Nasal Cartilages/chemistry , Nasal Cartilages/metabolism , Peptides/chemistry , Calcium, Dietary/analysis
2.
Development ; 142(7): 1357-67, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25742798

ABSTRACT

Bone morphogenetic protein (BMP) signaling plays many roles in skull morphogenesis. We have previously reported that enhanced BMP signaling through the BMP type IA receptor (BMPR1A) in cranial neural crest cells causes craniosynostosis during postnatal development. Additionally, we observed that 55% of Bmpr1a mutant mice show neonatal lethality characterized by a distended gastrointestinal tract. Here, we show that severely affected mutants exhibit defective nasal cartilage, failure of fusion between the nasal septum and the secondary palate, and higher levels of phosphorylated SMAD1 and SMAD5 in the nasal tissue. TUNEL demonstrated an increase in apoptosis in both condensing mesenchymal tissues and cartilage of the nasal region in mutants. The levels of p53 (TRP53) tumor suppressor protein were also increased in the same tissue. Injection of pifithrin-α, a chemical inhibitor of p53, into pregnant mice prevented neonatal lethality while concomitantly reducing apoptosis in nasal cartilage primordia, suggesting that enhanced BMP signaling induces p53-mediated apoptosis in the nasal cartilage. The expression of Bax and caspase 3, downstream targets of p53, was increased in the mutants; however, the p53 expression level was unchanged. It has been reported that MDM2 interacts with p53 to promote degradation. We found that the amount of MDM2-p53 complex was decreased in all mutants, and the most severely affected mutants had the largest decrease. Our previous finding that the BMP signaling component SMAD1 prevents MDM2-mediated p53 degradation coupled with our new data indicate that augmented BMP signaling induces p53-mediated apoptosis by prevention of p53 degradation in developing nasal cartilage. Thus, an appropriate level of BMP signaling is required for proper craniofacial morphogenesis.


Subject(s)
Apoptosis , Bone Morphogenetic Proteins/metabolism , Morphogenesis , Nasal Cartilages/embryology , Neural Crest/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Benzothiazoles/pharmacology , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Chondrogenesis/drug effects , Embryo Loss/metabolism , Embryo Loss/pathology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Fibroblast Growth Factors/metabolism , Integrases/metabolism , Mesoderm/drug effects , Mesoderm/embryology , Mesoderm/pathology , Mice , Morphogenesis/drug effects , Mutation/genetics , Nasal Cartilages/abnormalities , Nasal Cartilages/metabolism , Nasal Cartilages/pathology , Nasal Mucosa/metabolism , Neural Crest/drug effects , Neural Crest/embryology , Nose/embryology , Protein Binding/drug effects , Proteolysis/drug effects , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Toluene/analogs & derivatives , Toluene/pharmacology
3.
Oral Dis ; 24(1-2): 78-83, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29480643

ABSTRACT

Craniofacial development is a delicate process that involves complex interactions among cells of multiple developmental origins, their migration, proliferation, and differentiation. Tissue morphogenesis of the craniofacial skeleton depends on genetic and environmental factors, and on specific signaling pathways, which are still not well understood. Developmental defects of the midface caused by the absence, delays, or premature fusion of nasal and maxillary prominences vary in severity; leading to clefts, hypoplasias, and midline expansion. In the current review, we focus on the importance of the chondrocranium in craniofacial growth and how its impaired development leads to midface hypoplasia. More importantly, we reported how Matrix Gla protein (MGP), a potent inhibitor of extracellular matrix mineralization, facilitates midface development by preventing ectopic calcification of the nasal septum. In fact, MGP may act as a common link in multiple developmental pathologies all showing midface hypoplasia caused by abnormal cartilage calcification. This brief review discusses the gap in knowledge in the field, raises pertinent questions, which remain unanswered, and sheds light on the future research directions.


Subject(s)
Calcinosis/metabolism , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Face/abnormalities , Facial Bones/growth & development , Maxillofacial Development , Nasal Cartilages/growth & development , Calcinosis/congenital , Extracellular Matrix/metabolism , Facial Bones/abnormalities , Humans , Nasal Cartilages/abnormalities , Nasal Cartilages/metabolism , Matrix Gla Protein
4.
Eur Arch Otorhinolaryngol ; 274(9): 3503-3512, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28669050

ABSTRACT

The aim of this study was to analyze the role of Ki-67, p53, and the "aberrant p53 pattern" in squamous cell carcinomas of the nasal vestibule. Patients between 1995 and 2014 were included. Baseline characteristics and outcome were analyzed with respect to immunohistochemical staining of Ki-67 and p53. "Aberrant p53 pattern" was represented by a moderate or strong staining of at least 60% of the tumor cells or a complete absence of immunoreactivity. Forty-six patients were included of whom 31 (67.4%) were available for Ki-67 and 32 (69.9%) for p53 immunohistochemistry. The "aberrant pattern" of p53 was present in 50% of the patients. While immunoreactivity for both Ki-67 and p53 was not related to each other or outcome, the "aberrant p53 pattern" was associated with a worse disease-free survival (p = 0.014). The "aberrant p53 pattern" is a negative prognostic factor in squamous cell carcinoma of the nasal vestibule and might enable a patient-tailored treatment.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Nose Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/mortality , Disease-Free Survival , Female , Humans , Immunohistochemistry , Male , Middle Aged , Nasal Cartilages/metabolism , Nasal Cartilages/pathology , Neoplasm Staging , Nose Neoplasms/diagnosis , Nose Neoplasms/mortality , Prognosis , Survival Rate/trends , Switzerland/epidemiology
5.
Glycobiology ; 25(5): 557-69, 2015 May.
Article in English | MEDLINE | ID: mdl-25533443

ABSTRACT

Epiphycan (EPY) from salmon nasal cartilage has a glycosaminoglycan (GAG) domain that is heavily modified by chondroitin 4-sulfate and chondroitin 6-sulfate. The functional role of the GAG domain has not been investigated. The interaction of EPY with collagen was examined in vitro using surface plasmon resonance analysis. EPY was found to bind to type I collagen via clustered chondroitin sulfate (CS), while a single chain of CS was unable to bind. Types I, III, VII, VIII and X collagen showed high binding affinity with EPY, whereas types II, IV, V, VI and IX showed low binding affinities. Chemical modification of lysine residues in collagen decreased the affinity with the clustered CS. These results suggest that lysine residues of collagen are involved in the interaction with the clustered CS, and the difference in lysine modification defines the binding affinity to EPY. The clustered CS was also involved in an inter-saccharide interaction, and formed self-associated EPY. CS of EPY promoted fibril formation of type I collagen.


Subject(s)
Chondroitin Sulfates/metabolism , Collagen/metabolism , Nasal Cartilages/metabolism , Proteoglycans/metabolism , Animals , Protein Binding , Salmon
6.
Eur Arch Otorhinolaryngol ; 272(7): 1667-71, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25129374

ABSTRACT

Any abnormality of collagen may affect the tissues with higher collagen content, e.g., joints, heart valves, and great arteries. Mitral valve prolapse (MVP) is a characteristic of generalized collagen abnormality. Nasal septum (NS) is constituted by osseous and cartilaginous septums that are highly rich in collagen. We evaluated the co-existence of deviation of NS (DNS) in patients with MVP. We retrospectively evaluated the recordings of echocardiographic and nasal examinations of subjects with MVP and DNS. We analyzed the features of MVP and anatomical classification of DNS among subjects. Totally, 74 patients with DNS and 38 subjects with normal nasal passage were enrolled to the study. Presence of MVP was significantly higher in patients with DNS compared to normal subjects (63 vs 26%, p < 0.001). Prolapse of anterior, posterior and both leaflets was higher in patients with DNS. Thickness of anterior mitral leaflet was significantly increased in patients with DNS (3.57 ± 0.68 vs 4.59 ± 1.1 mm, p < 0.001) compared to normal subjects. Type I, II, and III, IV DNS were higher in frequency in patients with MVP while type V and VI were higher in normal subjects. DNS is highly co-existent with MVP and increased thickness of mitral anterior leaflet. Generalized abnormality of collagen which is the main component of mitral valves and nasal septum may be accounted for co-existence of MVP and DNS. Also co-existence of them may exaggerate the symptoms of patients with MVP due to limited airflow through the nasal passage.


Subject(s)
Mitral Valve Prolapse , Nasal Septum/pathology , Nose Deformities, Acquired , Nose/abnormalities , Adult , Collagen/metabolism , Echocardiography/methods , Female , Humans , Male , Middle Aged , Mitral Valve/pathology , Mitral Valve Prolapse/complications , Mitral Valve Prolapse/diagnosis , Nasal Cartilages/metabolism , Nasal Cartilages/pathology , Nose Deformities, Acquired/complications , Nose Deformities, Acquired/diagnosis , Retrospective Studies
7.
Arthritis Rheum ; 65(7): 1822-30, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23575963

ABSTRACT

OBJECTIVE: To examine the ability of a broad-spectrum histone deacetylase (HDAC) inhibitor to protect cartilage in vivo, and to explore the effects of class-selective HDAC inhibitors and small interfering RNA (siRNA)-induced knockdown of HDACs on metalloproteinase expression and cartilage degradation in vitro. METHODS: A destabilization of the medial meniscus (DMM) model was used to assess the in vivo activity of the HDAC inhibitor trichostatin A (TSA). Human articular chondrocytes (HACs) and SW-1353 chondrosarcoma cells were treated with cytokines and TSA, valproic acid, MS-275, or siRNA, and quantitative reverse transcription-polymerase chain reaction was performed to determine the effect of treatment on metalloproteinase expression. HDAC inhibitor activity was detected by Western blotting. A bovine nasal cartilage (BNC) explant assay was performed to measure cartilage resorption in vitro. RESULTS: Systemically administered TSA protected cartilage in the DMM model. TSA, valproic acid, and MS-275 repressed cytokine-induced MMP1 and MMP13 expression in HACs. Knockdown of each class I HDAC diminished interleukin-1-induced MMP13 expression. All of the HDAC inhibitors prevented degradation of BNC, in which TSA and MS-275 repressed cytokine-induced MMP expression. CONCLUSION: Inhibition of class I HDACs (HDAC-1, HDAC-2, HDAC-3) by MS-275 or by specific depletion of HDACs is capable of repressing cytokine-induced metalloproteinase expression in cartilage cells and BNC explants, resulting in inhibition of cartilage resorption. These observations indicate that specific inhibition of class I HDACs is a possible therapeutic strategy in the arthritides.


Subject(s)
Benzamides/pharmacology , Chondrocytes/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Metalloproteases/drug effects , Nasal Cartilages/drug effects , Osteoarthritis, Knee , Pyridines/pharmacology , Animals , Cattle , Cell Line, Tumor , Cells, Cultured , Chondrocytes/metabolism , Disease Models, Animal , Histones/drug effects , Histones/metabolism , Humans , Metalloproteases/metabolism , Mice , Mice, Inbred C57BL , Nasal Cartilages/metabolism , RNA, Small Interfering/pharmacology , Tubulin/drug effects , Tubulin/metabolism
8.
J Dent Res ; 103(3): 308-317, 2024 03.
Article in English | MEDLINE | ID: mdl-38234039

ABSTRACT

Neural crest cells (NCC) arise from the dorsal margin of the neural plate border and comprise a unique cell population that migrates to and creates the craniofacial region. Although factors including Shh, Fgf8, and bone morphogenetic proteins have been shown to regulate these biological events, the role of parathyroid hormone 1 receptor (Pth1r) has been less studied. We generated an NCC-specific mouse model for Pth1r and researched gene expression, function, and interaction focusing on nasal cartilage framework and midfacial development. Wnt1-Cre;Pth1rfl/fl;Tomatofl/+ mice had perinatal lethality, but we observed short snout and jaws, tongue protrusion, reduced NCC-derived cranial length, increased mineralization in nasal septum and hyoid bones, and less bone mineralization at interfrontal suture in mutants at E18.5. Importantly, the mutant nasal septum and turbinate cartilage histologically revealed gradual, premature accelerated hypertrophic differentiation. We then studied the underlying molecular mechanisms by performing RNA seq analysis and unexpectedly found that expression of Ihh and related signaling molecules was enhanced in mutant nasomaxillary tissues. To see if Pth1r and Ihh signaling are associated, we generated a Wnt1-Cre; Ihhfl/fl;Pth1rfl/fl;Tomatofl/+ (DKO) mouse and compared the phenotypes to those of each single knockout mouse: Wnt1-Cre; Ihhfl/fl;Pth1rfl/+;Tomatofl/+ (Ihh-CKO) and Wnt1-Cre;Ihhfl/+;Pth1rfl/fl;Tomatofl/+ (Pth1r-CKO). Ihh-CKO mice displayed a milder effect. Of note, the excessive hypertrophic conversion of the nasal cartilage framework observed in Pth1r-CKO was somewhat rescued DKO embryos. Further, a half cAMP responsive element and the 4 similar sequences containing 2 mismatches were identified from the promoter to the first intron in Ihh gene. Gli1-CreERT2;Pth1rfl/fl;Tomatofl/+, a Pth1r-deficient model targeted in hedgehog responsive cells, demonstrated the enlarged hypertrophic layer and significantly more Tomato-positive chondrocytes accumulated in the nasal septum and ethmoidal endochondral ossification. Collectively, the data suggest a relevant Pth1r/Ihh interaction. Our findings obtained from novel mouse models for Pth1r signaling illuminate previously unknown aspects in craniofacial biology and development.


Subject(s)
Nasal Cartilages , Neural Crest , Receptor, Parathyroid Hormone, Type 1 , Animals , Mice , Cell Differentiation , Chondrocytes/metabolism , Hedgehog Proteins/metabolism , Mice, Knockout , Nasal Cartilages/metabolism , Skull , Receptor, Parathyroid Hormone, Type 1/metabolism
9.
Int J Biol Macromol ; 277(Pt 4): 134501, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39111483

ABSTRACT

This study employs an optimized and environmentally friendly method to extract and purify chondroitin sulfate (CS) from bovine nasal cartilage using enzymatic hydrolysis, ethanol precipitation, and DEAE Sepharose Fast Flow column chromatography. The extracted CS, representing 44.67 % ± 0.0016 of the cartilage, has a molecular weight of 7.62 kDa. Characterization through UV, FT-IR, NMR spectroscopy, and 2-aminoacridone derivatization HPLC revealed a high content of sulfated disaccharides, particularly ΔDi4S (73.59 %) and ΔDi6S (20.61 %). Interaction studies with bovine serum albumin (BSA) using fluorescence spectroscopy and molecular docking confirmed a high-affinity, static quenching interaction with a single binding site, primarily mediated by van der Waals forces and hydrogen bonding. The interaction did not significantly alter the polarity or hydrophobicity of BSA aromatic amino acids. These findings provide a strong foundation for exploring the application of CS in tissue engineering and drug delivery systems, leveraging its unique interaction with BSA for targeted delivery and enhanced efficacy.


Subject(s)
Chondroitin Sulfates , Nasal Cartilages , Serum Albumin, Bovine , Animals , Cattle , Chondroitin Sulfates/chemistry , Molecular Docking Simulation , Molecular Weight , Nasal Cartilages/chemistry , Nasal Cartilages/metabolism , Protein Binding , Serum Albumin, Bovine/chemistry
10.
Cell Biochem Funct ; 31(3): 214-27, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23001693

ABSTRACT

The capacity of cartilage self-regeneration is considered to be limited. Joint injuries often evolve in the development of chronic wounds on the cartilage surface. Such lesions are associated with articular cartilage degeneration and osteoarthritis. Re-establishing a correct micro/macro-environment into damaged joints could stop or prevent the degenerative processes. This study investigated the effect of polydeoxyribonucleotides (PDRNs) on cartilage degradation in vitro and on cartilage extracted cells. The activities of matrix metalloproteinases 2 and 9 were measured in PDRN-treated cells and in controls at days 0 and 30 of culture. Human nasal cartilage explants were cultured, and the degree of proteoglycan degradation was assessed by measuring the amount of glycosaminoglycans released into the culture medium. The PDRN properties compared with controls were tested on cartilage tissues to evaluate deposition of extracellular matrix. Chondrocytes treated with PDRNs showed a physiological deposition of extracellular matrix (aggrecan and type II collagen: Western blot, IFA, fluorescence activated cell sorting, Alcian blue and safranin O staining). PDRNs were able to inhibit proteoglycan degradation in cartilage explants. The activities of matrix metalloproteinases 2 and 9 were reduced in all PDRN-treated samples. Our results indicate that PDRNs are suitable for a long-term cultivation of in vitro cartilage and have therapeutic effects on chondrocytes by protecting cartilage.


Subject(s)
Nasal Cartilages/drug effects , Polydeoxyribonucleotides/pharmacology , Protective Agents/pharmacology , Adult , Aggrecans/metabolism , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Collagen Type II/metabolism , Extracellular Matrix/metabolism , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Nasal Cartilages/cytology , Nasal Cartilages/metabolism
11.
Ann Rheum Dis ; 71(3): 455-62, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22072016

ABSTRACT

OBJECTIVES: To investigate the effect of leptin on cartilage destruction. METHODS: Collagen release was assessed in bovine cartilage explant cultures, while collagenolytic and gelatinolytic activities in culture supernatants were determined by bioassay and gelatin zymography. The expression of matrix metalloproteinases (MMP) was analysed by real-time RT-PCR. Signalling pathway activation was studied by immunoblotting. Leptin levels in cultured osteoarthritic joint infrapatellar fat pad or peri-enthesal deposit supernatants were measured by immunoassay. RESULTS: Leptin, either alone or in synergy with IL-1, significantly induced collagen release from bovine cartilage by upregulating collagenolytic and gelatinolytic activity. In chondrocytes, leptin induced MMP1 and MMP13 expression with a concomitant activation of STAT1, STAT3, STAT5, MAPK (JNK, Erk, p38), Akt and NF-κB signalling pathways. Selective inhibitor blockade of PI3K, p38, Erk and Akt pathways significantly reduced MMP1 and MMP13 expression in chondrocytes, and reduced cartilage collagen release induced by leptin or leptin plus IL-1. JNK inhibition had no effect on leptin-induced MMP13 expression or leptin plus IL-1-induced cartilage collagen release. Conditioned media from cultured white adipose tissue (WAT) from osteoarthritis knee joint fat pads contained leptin, induced cartilage collagen release and increased MMP1 and MMP13 expression in chondrocytes; the latter being partly blocked with an anti-leptin antibody. CONCLUSIONS: Leptin acts as a pro-inflammatory adipokine with a catabolic role on cartilage metabolism via the upregulation of proteolytic enzymes and acts synergistically with other pro-inflammatory stimuli. This suggests that the infrapatellar fat pad and other WAT in arthritic joints are local producers of leptin, which may contribute to the inflammatory and degenerative processes in cartilage catabolism, providing a mechanistic link between obesity and osteoarthritis.


Subject(s)
Adipose Tissue, White/metabolism , Cartilage, Articular/metabolism , Leptin/physiology , Matrix Metalloproteinases/physiology , Animals , Cattle , Cells, Cultured , Collagen/metabolism , Collagenases/biosynthesis , Collagenases/genetics , Culture Media, Conditioned , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Humans , Inflammation Mediators/pharmacology , Leptin/biosynthesis , Leptin/pharmacology , Matrix Metalloproteinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Nasal Cartilages/drug effects , Nasal Cartilages/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tissue Culture Techniques
12.
MAGMA ; 25(4): 297-304, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22071581

ABSTRACT

OBJECTIVE: To study the orientational dependencies of T(2) and T(1ρ) in native and trypsin-degraded bovine nasal cartilage, with and without the presence of 1 mM Gd-DTPA(2-). MATERIALS AND METHODS: Sixteen specimens were prepared in two orthogonal fibril directions (parallel and perpendicular), treated using different protocols (native, Gd treated, trypsin-treated, and combination), and imaged using µMRI at 0° and 55° (the magic angle) fibril orientations with respect to the magnetic field B(0). Two-dimensional (2D) T(2) and T(1ρ) images were then calculated quantitatively. RESULTS: Without Gd, native perpendicular tissues demonstrated significant T(1ρ) dispersion (including T(2) at the zero spin-lock field) at 0° and less dispersion at 55°, while native parallel specimens exhibited smaller T(1ρ) dispersion at both 0° and 55°. Trypsin degradation caused a minimum 50% increase in T(1ρ). With Gd, trypsin degradation caused significant reduction in T(1ρ) values up to 60%. CONCLUSION: The collagen orientation in nasal cartilage can influence T(2) and T(1ρ) MRI of cartilage. Without Gd, T(1ρ) was sensitive to the proteoglycan content and its sensitivity was nearly constant regardless of fibril orientation. In comparison, the T(2) sensitivity to proteoglycan was dependant upon fibril orientation, i.e., more sensitive at 55° than 0°. When Gd ions were present, both T(2) and T(1ρ) became insensitive to the proteoglycan content.


Subject(s)
Magnetic Resonance Imaging/methods , Nasal Cartilages/anatomy & histology , Nasal Cartilages/metabolism , Animals , Cattle , Collagen/metabolism , Contrast Media , Gadolinium DTPA , Proteoglycans/metabolism , Trypsin
13.
Magn Reson Med ; 65(2): 377-84, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21264931

ABSTRACT

Association of MR parameters with cartilage matrix components remains an area of ongoing investigation. Multiexponential analysis of nonlocalized transverse relaxation data has previously been used to quantify water compartments associated with matrix macromolecules in cartilage. We extend this to mapping the proteoglycan (PG)-bound water fraction in cartilage, using mature and young bovine nasal cartilage model systems, toward the goal of matrix component-specific imaging. PG-bound water fraction from mature and young bovine nasal cartilage was 0.31 ± 0.04 and 0.22 ± 0.06, respectively, in agreement with biochemically derived PG content and PG-to-water weight ratios. Fourier transform infrared imaging spectroscopic-derived PG maps normalized by water content (IR-PG(ww) ) showed spatial correspondence with PG-bound water fraction maps. Extensive simulation analysis demonstrated that the accuracy and precision of our determination of PG-bound water fraction was within 2%, which is well-within the observed tissue differences. Our results demonstrate the feasibility of performing imaging-based multiexponential analysis of transverse relaxation data to map PG in cartilage.


Subject(s)
Cartilage, Articular/metabolism , Magnetic Resonance Spectroscopy/methods , Nasal Cartilages/metabolism , Proteoglycans/analysis , Animals , Cattle , In Vitro Techniques , Patella , Spectroscopy, Fourier Transform Infrared
14.
NMR Biomed ; 24(10): 1286-94, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21465593

ABSTRACT

The noninvasive early detection of specific matrix alterations in degenerative cartilage disease would be of substantial use in basic science studies and clinically, but remains an elusive goal. Recently developed MRI methods exhibit some specificity, but require contrast agents or nonstandard pulse sequences and hardware. We present a multiexponential approach which does not require contrast agents or specialized hardware, and uses a standard multiple-echo spin-echo sequence. Experiments were performed on tissue models of degenerative cartilage using enzymes with distinct actions. MR results were validated using histologic, biochemical and infrared spectroscopic analyses. The sulfated glycosaminoglycan per dry weight (dw) in bovine nasal cartilage was 0.72 ± 0.06 mg/mg dw and was reduced through chondroitinase AC and collagenase digestion to 0.56 ± 0.12 and 0.58 ± 0.13 mg/mg dw, respectively. Multiexponential analysis of data obtained at 9.4 T permitted the identification of tissue compartments assigned to the proteoglycan component of the matrix and to bulk water. Enzymatic treatment resulted in a significant reduction in the ratio of proteoglycan-bound to free water from 0.13 ± 0.02 in control cartilage to 0.03 ± 0.02 and 0.05 ± 0.06 under chondroitinase AC and collagenase treatment, respectively. As expected, monoexponential T(2) increased with both degradation protocols, but without further specificity to the nature of the degradation. An important eventual extension of this approach may be to map articular cartilage degeneration in the clinical setting. As an initial step towards this, localized multiexponential T(2) analysis was performed on control and trypsin treated excised bovine patella. The results obtained on this articular cartilage sample were readily interpretable in terms of proteoglycan-associated and relatively free water compartments. In potential clinical applications, signal-to-noise ratio constraints will define the threshold for the detection of macromolecular compartment changes at a given spatial scale. The multiexponential approach has potential application to the early detection of cartilage degradation with the use of appropriate pulse parameters under high signal-to-noise ratio conditions.


Subject(s)
Cartilage/metabolism , Cartilage/pathology , Extracellular Matrix/metabolism , Magnetic Resonance Imaging/methods , Alcian Blue/metabolism , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cattle , Computer Simulation , Glycosaminoglycans/metabolism , Nasal Cartilages/metabolism , Nasal Cartilages/pathology , Patella/metabolism , Patella/pathology , Spectroscopy, Fourier Transform Infrared
15.
PLoS Genet ; 4(12): e1000308, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19096511

ABSTRACT

Cartilage and bone are formed into a remarkable range of shapes and sizes that underlie many anatomical adaptations to different lifestyles in vertebrates. Although the morphological blueprints for individual cartilage and bony structures must somehow be encoded in the genome, we currently know little about the detailed genomic mechanisms that direct precise growth patterns for particular bones. We have carried out large-scale enhancer surveys to identify the regulatory architecture controlling developmental expression of the mouse Bmp5 gene, which encodes a secreted signaling molecule required for normal morphology of specific skeletal features. Although Bmp5 is expressed in many skeletal precursors, different enhancers control expression in individual bones. Remarkably, we show here that different enhancers also exist for highly restricted spatial subdomains along the surface of individual skeletal structures, including ribs and nasal cartilages. Transgenic, null, and regulatory mutations confirm that these anatomy-specific sequences are sufficient to trigger local changes in skeletal morphology and are required for establishing normal growth rates on separate bone surfaces. Our findings suggest that individual bones are composite structures whose detailed growth patterns are built from many smaller lineage and gene expression domains. Individual enhancers in BMP genes provide a genomic mechanism for controlling precise growth domains in particular cartilages and bones, making it possible to separately regulate skeletal anatomy at highly specific locations in the body.


Subject(s)
Bone Development , Bone Morphogenetic Protein 5/genetics , Nasal Cartilages/growth & development , Regulatory Sequences, Nucleic Acid , Ribs/growth & development , Animals , Bone Morphogenetic Protein 5/chemistry , Bone Morphogenetic Protein 5/metabolism , Enhancer Elements, Genetic , Gene Expression , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nasal Cartilages/embryology , Nasal Cartilages/metabolism , Protein Structure, Tertiary , Ribs/anatomy & histology , Ribs/embryology , Ribs/metabolism , Signal Transduction
16.
Ann Rheum Dis ; 69(12): 2189-98, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20693272

ABSTRACT

OBJECTIVE: To investigate if statins prevent cartilage degradation and the production of collagenases and gelatinases in bovine nasal and human articular cartilage after proinflammatory cytokine stimulation. METHODS: In a cartilage degradation model, the effects of several statins were assessed by measuring proteoglycan degradation and collagen degradation, while collagenolytic and gelatinolytic activity in culture supernatants were determined by collagen bioassay and gelatin zymography. The production of matrix metalloproteinases (MMPs) in cartilage and chondrocytes were analysed by real-time reverse transcriptase PCR and immunoassay. Cytokine-induced signalling pathway activation was studied by immunoblotting. RESULTS: Simvastatin and mevastatin significantly inhibited interleukin 1 (IL-1)+oncostatin M (OSM)-induced collagen degradation; this was accompanied with a marked decrease in collagenase and gelatinase activity from bovine nasal cartilage. The cholesterol pathway intermediate mevalonic acid reversed the simvastatin-mediated protection of cartilage degradation, and the expression and production of collagenase (MMP-1 and MMP-13) and gelatinase (MMP-2 and MMP-9). Statins also significantly decreased MMP-1 and MMP-13 expression in human articular cartilage and chondrocytes stimulated with IL-1+OSM, and blocked the activation of critical proinflammatory signalling pathways required for MMP expression. The loss of the isoprenoid intermediate geranylgeranyl pyrophosphate due to statin treatment accounted for the inhibition of MMP expression and signalling pathway activation. CONCLUSIONS: This study shows, for the first time, that lipophilic statins are able to block cartilage collagen breakdown induced by proinflammatory cytokines, by downregulating key cartilage-degrading enzymes. This demonstrates a possible therapeutic role for statins in acting as anti-inflammatory agents and in protecting cartilage from damage in joint diseases.


Subject(s)
Cartilage, Articular/drug effects , Collagen/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Matrix Metalloproteinases/physiology , Nasal Cartilages/drug effects , Animals , Cartilage, Articular/metabolism , Cattle , Cells, Cultured , Collagenases/biosynthesis , Down-Regulation/drug effects , Gelatinases/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Humans , Interleukin-1alpha/pharmacology , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Matrix Metalloproteinases/genetics , Mevalonic Acid/pharmacology , Nasal Cartilages/metabolism , Oncostatin M/pharmacology , Signal Transduction/drug effects , Simvastatin/antagonists & inhibitors , Simvastatin/pharmacology , Terpenes/metabolism , Tissue Culture Techniques
17.
Rheumatology (Oxford) ; 48(10): 1208-12, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19690126

ABSTRACT

OBJECTIVE: To investigate the effect of SSZ on the release of GAG and collagen fragments from bovine nasal cartilage and MMP and ADAMTS (a disintegrin and metalloproteinase domain with thrombospondin motifs) proteinases from human articular chondrocytes (HACs) stimulated with IL-1alpha and oncostatin M (OSM). METHODS: SSZ was added to bovine nasal explant cultures stimulated to resorb with IL-1alpha and OSM, and the release of GAG and collagen has been determined. Collagenolytic activity was measured using the radio-labelled collagen bioassay. HACs were treated with IL-1alpha and OSM with and without SSZ, and MMP-1 and -13 and ADAMTS-4 and -5 were measured for protein and gene expression by ELISA and RT-PCR, respectively. RESULTS: SSZ blocked GAG and collagen fragment release from bovine cartilage, and reduced active and total collagenase activity in a dose-dependent manner. SSZ transcriptionally blocked MMP-1, -13 and ADAMTS-4, and reduced the protein levels of MMP-1 and -13 in a dose-dependent manner following stimulation of HACs with IL-1alpha and OSM. CONCLUSION: This study shows for the first time that SSZ blocks release of proteoglycan and collagen fragments from resorbing cartilage and lowers the levels of proteoglycan and collagen-degrading enzymes. These results indicate that in addition to acting as an anti-inflammatory agent, SSZ may have a therapeutic role in protecting cartilage from damage in OA.


Subject(s)
Antirheumatic Agents/pharmacology , Collagen/metabolism , Hyaline Cartilage/drug effects , Proteoglycans/metabolism , Sulfasalazine/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cattle , Cells, Cultured , Culture Media, Conditioned , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Evaluation, Preclinical/methods , Gene Expression Regulation, Enzymologic/drug effects , Humans , Hyaline Cartilage/metabolism , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1alpha/pharmacology , Metalloproteases/biosynthesis , Nasal Cartilages/drug effects , Nasal Cartilages/metabolism , Oncostatin M/antagonists & inhibitors , Oncostatin M/pharmacology , Osteoarthritis, Knee/metabolism
18.
Acta Biomater ; 88: 42-56, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30794988

ABSTRACT

Nasal cartilage pathologies are common; for example, up to 80% of people are afflicted by deviated nasal septum conditions. Because cartilage provides the supportive framework of the nose, afflicted patients suffer low quality of life. To correct pathologies, graft cartilage is often required. Grafts are currently sourced from the patient's septum, ear, or rib. However, their use yields donor site morbidity and is limited by tissue quantity and quality. Additionally, rhinoplasty revision rates exceed 15%, exacerbating the shortage of graft cartilage. Alternative grafts, such as irradiated allogeneic rib cartilage, are associated with complications. Tissue-engineered neocartilage holds promise to address the limitations of current grafts. The engineering design process may be used to create suitable graft tissues. This process begins by identifying the surgeon's needs. Second, nasal cartilages' properties must be understood to define engineering design criteria. Limited investigations have examined nasal cartilage properties; numerous additional studies need to be performed to examine topographical variations, for example. Third, tissue-engineering processes must be applied to achieve the engineering design criteria. Within the recent past, strategies have frequently utilized human septal chondrocytes. As autologous and allogeneic rib graft cartilage is used, its suitability as a cell source should also be examined. Fourth, quantitative verification of engineered neocartilage is critical to check for successful achievement of the engineering design criteria. Finally, following the FDA paradigm, engineered neocartilage must be orthotopically validated in animals. Together, these steps delineate a path to engineer functional nasal neocartilages that may, ultimately, be used to treat human patients. STATEMENT OF SIGNIFICANCE: Nasal cartilage pathologies are common and lead to greatly diminished quality of life. The ability to correct pathologies is limited by cartilage graft quality and quantity, as well as donor site morbidity and surgical complications, such as infection and resorption. Despite the significance of nasal cartilage pathologies and high rhinoplasty revision rates (15%), little characterization and tissue-engineering work has been performed compared to other cartilages, such as articular cartilage. Furthermore, most work is published in clinical journals, with little in biomedical engineering. Therefore, this review discusses what nasal cartilage properties are known, summarizes the current state of nasal cartilage tissue-engineering, and makes recommendations via the engineering design process toward engineering functional nasal neocartilage to address current limitations.


Subject(s)
Cartilage, Articular , Nasal Cartilages , Rhinoplasty , Tissue Engineering , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cartilage, Articular/transplantation , Humans , Nasal Cartilages/metabolism , Nasal Cartilages/pathology , Nasal Cartilages/transplantation , Quality of Life , Transplantation, Autologous
19.
J Biomed Mater Res A ; 107(4): 893-903, 2019 04.
Article in English | MEDLINE | ID: mdl-30650239

ABSTRACT

Nasal reconstruction remains a challenge for every reconstructive surgeon. Alloplastic implants are proposed to repair nasal cartilaginous defects but they are often associated with high rates of extrusion and infection and poor biocompatibility. In this context, a porous polymeric scaffold filled with an autologous cartilage gel would be advantageous. In this study, we evaluated the capacity of IEIK13 self-assembling peptide (SAP) to serve as support to form such cartilage gel. Human nasal chondrocytes (HNC) were first amplified with FGF-2 and insulin, and then redifferentiated in IEIK13 with BMP-2, insulin, and T3 (BIT). Our results demonstrate that IEIK13 fosters HNC growth and survival. HNC phenotype was assessed by RT-PCR analysis and neo-synthesized extracellular matrix was characterized by western blotting and immunohistochemistry analysis. BIT-treated cells embedded in IEIK13 displayed round morphology and expressed cartilage-specific markers such as type II and type IX collagens and aggrecan. In addition, we did not detect significant production of type I and type X collagens and gene products of dedifferentiated and hypertrophic chondrocytes that are unwanted in hyaline cartilage. The whole of these results indicates that the SAP IEIK13 represents a suitable support for hydrogel-based tissue engineering of nasal cartilage. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 893-903, 2019.


Subject(s)
Chondrocytes/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Hydrogels/chemistry , Nasal Cartilages/metabolism , Peptides/chemistry , Adult , Chondrocytes/cytology , Female , Humans , Male , Middle Aged , Nasal Cartilages/cytology
20.
Elife ; 72018 06 13.
Article in English | MEDLINE | ID: mdl-29897331

ABSTRACT

Facial shape is the basis for facial recognition and categorization. Facial features reflect the underlying geometry of the skeletal structures. Here, we reveal that cartilaginous nasal capsule (corresponding to upper jaw and face) is shaped by signals generated by neural structures: brain and olfactory epithelium. Brain-derived Sonic Hedgehog (SHH) enables the induction of nasal septum and posterior nasal capsule, whereas the formation of a capsule roof is controlled by signals from the olfactory epithelium. Unexpectedly, the cartilage of the nasal capsule turned out to be important for shaping membranous facial bones during development. This suggests that conserved neurosensory structures could benefit from protection and have evolved signals inducing cranial cartilages encasing them. Experiments with mutant mice revealed that the genomic regulatory regions controlling production of SHH in the nervous system contribute to facial cartilage morphogenesis, which might be a mechanism responsible for the adaptive evolution of animal faces and snouts.


Subject(s)
Brain/metabolism , Chondrocytes/metabolism , Hedgehog Proteins/genetics , Maxillofacial Development/genetics , Morphogenesis/genetics , Olfactory Mucosa/metabolism , Signal Transduction , Animals , Brain/drug effects , Brain/growth & development , Chondrocytes/cytology , Chondrocytes/drug effects , Collagen Type II/genetics , Collagen Type II/metabolism , Embryo, Mammalian , Face/anatomy & histology , Face/embryology , Facial Bones/cytology , Facial Bones/drug effects , Facial Bones/growth & development , Facial Bones/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Morphogenesis/drug effects , Mutagens/administration & dosage , Nasal Cartilages/cytology , Nasal Cartilages/drug effects , Nasal Cartilages/growth & development , Nasal Cartilages/metabolism , Olfactory Mucosa/cytology , Olfactory Mucosa/drug effects , Olfactory Mucosa/growth & development , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tamoxifen/administration & dosage , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish Proteins
SELECTION OF CITATIONS
SEARCH DETAIL