Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nat Mater ; 20(6): 892-903, 2021 06.
Article in English | MEDLINE | ID: mdl-33495631

ABSTRACT

The basement membrane (BM) is a special type of extracellular matrix and presents the major barrier cancer cells have to overcome multiple times to form metastases. Here we show that BM stiffness is a major determinant of metastases formation in several tissues and identify netrin-4 (Net4) as a key regulator of BM stiffness. Mechanistically, our biophysical and functional analyses in combination with mathematical simulations show that Net4 softens the mechanical properties of native BMs by opening laminin node complexes, decreasing cancer cell potential to transmigrate this barrier despite creating bigger pores. Our results therefore reveal that BM stiffness is dominant over pore size, and that the mechanical properties of 'normal' BMs determine metastases formation and patient survival independent of cancer-mediated alterations. Thus, identifying individual Net4 protein levels within native BMs in major metastatic organs may have the potential to define patient survival even before tumour formation. The ratio of Net4 to laminin molecules determines BM stiffness, such that the more Net4, the softer the BM, thereby decreasing cancer cell invasion activity.


Subject(s)
Basement Membrane/metabolism , Mechanical Phenomena , Neoplasm Metastasis , Biomechanical Phenomena , Cell Line, Tumor , Humans , Netrins/metabolism
2.
Int J Mol Sci ; 22(17)2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34502096

ABSTRACT

The potential of Fourier Transform infrared microspectroscopy (FTIR microspectroscopy) and multivariate analyses were applied for the classification of the frequency ranges responsible for the distribution changes of the main components of articular cartilage (AC) that occur during dietary ß-hydroxy-ß-methyl butyrate (HMB) supplementation. The FTIR imaging analysis of histological AC sections originating from 35-day old male piglets showed the change in the collagen and proteoglycan contents of the HMB-supplemented group compared to the control. The relative amount of collagen content in the superficial zone increased by more than 23% and in the middle zone by about 17%, while no changes in the deep zone were observed compared to the control group. Considering proteoglycans content, a significant increase was registered in the middle and deep zones, respectively; 62% and 52% compared to the control. AFM nanoindentation measurements collected from animals administered with HMB displayed an increase in AC tissue stiffness by detecting a higher value of Young's modulus in all investigated AC zones. We demonstrated that principal component analysis and artificial neural networks could be trained with spectral information to distinguish AC histological sections and the group under study accurately. This work may support the use and effectiveness of FTIR imaging combined with multivariate analyses as a quantitative alternative to traditional collagenous tissue-related histology.


Subject(s)
Cartilage, Articular/drug effects , Valerates/pharmacology , Animals , Cartilage, Articular/chemistry , Cartilage, Articular/metabolism , Collagen/metabolism , Dietary Supplements , Elastic Modulus , Male , Neural Networks, Computer , Principal Component Analysis , Proteoglycans/metabolism , Spectroscopy, Fourier Transform Infrared , Swine , Valerates/administration & dosage
3.
J Cell Mol Med ; 24(10): 5665-5674, 2020 05.
Article in English | MEDLINE | ID: mdl-32283567

ABSTRACT

In patients with osteomalacia, a defect in bone mineralization leads to changed characteristics of the bone surface. Considering that the properties of the surrounding matrix influence function and differentiation of cells, we aimed to investigate the effect of osteoidosis on differentiation and function of osteoclasts. Based on osteomalacic bone biopsies, a model for osteoidosis in vitro (OIV) was established. Peripheral blood mononuclear cells were differentiated to osteoclasts on mineralized surfaces (MS) as internal control and on OIV. We observed a significantly reduced number of osteoclasts and surface resorption on OIV. Atomic force microscopy revealed a significant effect of the altered degree of mineralization on surface mechanics and an unmasking of collagen fibres on the surface. Indeed, coating of MS with RGD peptides mimicked the resorption phenotype observed in OIV, suggesting that the altered differentiation of osteoclasts on OIV might be associated with an interaction of the cells with amino acid sequences of unmasked extracellular matrix proteins containing RGD sequences. Transcriptome analysis uncovered a strong significant up-regulation of transmembrane glycoprotein TROP2 in osteoclastic cultures on OIV. TROP2 expression on OIV was also confirmed on the protein level and found on the bone surface of patients with osteomalacia. Taken together, our results show a direct influence of the mineralization state of the extracellular matrix surface on differentiation and function of osteoclasts on this surface which may be important for the pathophysiology of osteomalacia and other bone disorders with changed ratio of osteoid to bone.


Subject(s)
Cell Differentiation , Osteoclasts/cytology , Osteoclasts/metabolism , Osteomalacia/etiology , Osteomalacia/metabolism , Biopsy , Bone and Bones/metabolism , Bone and Bones/pathology , Calcification, Physiologic , Cell Count , Cell Differentiation/genetics , Cells, Cultured , Extracellular Matrix/metabolism , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , Microscopy, Atomic Force , Osteoblasts/metabolism , Osteomalacia/pathology , Retrospective Studies , Transcriptome
4.
Int J Mol Sci ; 21(2)2020 Jan 19.
Article in English | MEDLINE | ID: mdl-31963938

ABSTRACT

Matrilins (MATN1, MATN2, MATN3 and MATN4) are adaptor proteins of the cartilage extracellular matrix (ECM), which bridge the collagen II and proteoglycan networks. In humans, dominant-negative mutations in MATN3 lead to various forms of mild chondrodysplasias. However, single or double matrilin knockout mice generated previously in our laboratory do not show an overt skeletal phenotype, suggesting compensation among the matrilin family members. The aim of our study was to establish a mouse line, which lacks all four matrilins and analyze the consequence of matrilin deficiency on endochondral bone formation and cartilage function. Matn1-4-/- mice were viable and fertile, and showed a lumbosacral transition phenotype characterized by the sacralization of the sixth lumbar vertebra. The development of the appendicular skeleton, the structure of the growth plate, chondrocyte differentiation, proliferation, and survival were normal in mutant mice. Biochemical analysis of knee cartilage demonstrated moderate alterations in the extractability of the binding partners of matrilins in Matn1-4-/- mice. Atomic force microscopy (AFM) revealed comparable compressive stiffness but higher collagen fiber diameters in the growth plate cartilage of quadruple mutant compared to wild-type mice. Importantly, Matn1-4-/- mice developed more severe spontaneous osteoarthritis at the age of 18 months, which was accompanied by changes in the biomechanical properties of the articular cartilage. Interestingly, Matn4-/- mice also developed age-associated osteoarthritis suggesting a crucial role of MATN4 in maintaining the stability of the articular cartilage. Collectively, our data provide evidence that matrilins are important to protect articular cartilage from deterioration and are involved in the specification of the vertebral column.


Subject(s)
Aging/genetics , Matrilin Proteins/genetics , Muscle, Skeletal/pathology , Osteoarthritis/pathology , Animals , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Disease Models, Animal , Female , Gene Knockout Techniques , Humans , Male , Mice , Mice, Knockout , Microscopy, Atomic Force , Osteoarthritis/genetics
5.
Ann Rheum Dis ; 76(2): 442-449, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27377816

ABSTRACT

OBJECTIVE: Interterritorial regions of articular cartilage matrix are rich in decorin, a small leucine-rich proteoglycan and important structural protein, also involved in many signalling events. Decorin sequesters transforming growth factor ß (TGFß), thereby regulating its activity. Here, we analysed whether increased bioavailability of TGFß in decorin-deficient (Dcn-/-) cartilage leads to changes in biomechanical properties and resistance to osteoarthritis (OA). METHODS: Unchallenged knee cartilage was analysed by atomic force microscopy (AFM) and immunohistochemistry. Active transforming growth factor ß-1 (TGFß1) content within cultured chondrocyte supernatants was measured by ELISA. Quantitative real-time (RT)-PCR was used to analyse mRNA expression of glycosaminoglycan (GAG)-modifying enzymes in C28/I2 cells following TGFß1 treatment. In addition, OA was induced in Dcn-/- and wild-type (WT) mice via forced exercise on a treadmill. RESULTS: AFM analysis revealed a strikingly higher compressive stiffness in Dcn-/- than in WT cartilage. This was accompanied by increased negative charge and enhanced sulfation of GAG chains, but not by alterations in the levels of collagens or proteoglycan core proteins. In addition, decorin-deficient chondrocytes were shown to release more active TGFß1. Increased TGFß signalling led to enhanced Chst11 sulfotransferase expression inducing an increased negative charge density of cartilage matrix. These negative charges might attract more water resulting in augmented compressive stiffness of the tissue. Therefore, decorin-deficient mice developed significantly less OA after forced exercise than WT mice. CONCLUSIONS: Our study demonstrates that the disruption of decorin-restricted TGFß signalling leads to higher stiffness of articular cartilage matrix, rendering joints more resistant to OA. Therefore, the loss of an important structural component can improve cartilage homeostasis.


Subject(s)
Arthritis, Experimental/genetics , Cartilage, Articular/metabolism , Decorin/genetics , Osteoarthritis/genetics , Physical Conditioning, Animal/methods , RNA, Messenger/metabolism , Transforming Growth Factor beta/metabolism , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/metabolism , Biomechanical Phenomena , Decorin/metabolism , Enzyme-Linked Immunosorbent Assay , Glycosaminoglycans/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Atomic Force , Osteoarthritis/etiology , Osteoarthritis/metabolism , Physical Conditioning, Animal/adverse effects , RNA, Messenger/drug effects , Real-Time Polymerase Chain Reaction , Transforming Growth Factor beta/pharmacology
6.
J Biol Chem ; 290(27): 16964-78, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-26001784

ABSTRACT

Collagen prolyl 4-hydroxylases (C-P4H-I, C-P4H-II, and C-P4H-III) catalyze formation of 4-hydroxyproline residues required to form triple-helical collagen molecules. Vertebrate C-P4Hs are α2ß2 tetramers differing in their catalytic α subunits. C-P4H-I is the major isoenzyme in most cells, and inactivation of its catalytic subunit (P4ha1(-/-)) leads to embryonic lethality in mouse, whereas P4ha1(+/-) mice have no abnormalities. To study the role of C-P4H-II, which predominates in chondrocytes, we generated P4ha2(-/-) mice. Surprisingly, they had no apparent phenotypic abnormalities. To assess possible functional complementarity, we established P4ha1(+/-);P4ha2(-/-) mice. They were smaller than their littermates, had moderate chondrodysplasia, and developed kyphosis. A transient inner cell death phenotype was detected in their developing growth plates. The columnar arrangement of proliferative chondrocytes was impaired, the amount of 4-hydroxyproline and the Tm of collagen II were reduced, and the extracellular matrix was softer in the growth plates of newborn P4ha1(+/-);P4ha2(-/-) mice. No signs of uncompensated ER stress were detected in the mutant growth plate chondrocytes. Some of these defects were also found in P4ha2(-/-) mice, although in a much milder form. Our data show that C-P4H-I can to a large extent compensate for the lack of C-P4H-II in proper endochondral bone development, but their combined partial and complete inactivation, respectively, leads to biomechanically impaired extracellular matrix, moderate chondrodysplasia, and kyphosis. Our mouse data suggest that inactivating mutations in human P4HA2 are not likely to lead to skeletal disorders, and a simultaneous decrease in P4HA1 function would most probably be required to generate such a disease phenotype.


Subject(s)
Chondrocytes/enzymology , Extracellular Matrix/metabolism , Osteochondrodysplasias/enzymology , Procollagen-Proline Dioxygenase/deficiency , Animals , Apoptosis , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Collagen/biosynthesis , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , Osteochondrodysplasias/embryology , Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Osteochondrodysplasias/physiopathology , Procollagen-Proline Dioxygenase/genetics
7.
Front Bioeng Biotechnol ; 11: 1128587, 2023.
Article in English | MEDLINE | ID: mdl-36937743

ABSTRACT

Aggrecan (ACAN) is localized in the intervertebral disc (IVD) in unique compartment-specific patterns where it contributes to the tissue structure and mechanical function together with collagens. The extracellular matrix (ECM) of the IVD undergoes degenerative changes during aging, misuse or trauma, which inevitably alter the biochemical and biomechanical properties of the tissue. A deeper understanding of these processes can be achieved in genetically engineered mouse models, taking into account the multifaceted aspects of IVD development. In this study, we generated aggrecan insertion mutant mice (Acan iE5/iE5 ) by interrupting exon 5 coding for the G1 domain of ACAN, and analyzed the morphological and mechanical properties of the different IVD compartments during embryonic development. Western blotting using an antibody against the total core protein failed to detect ACAN in cartilage extracts, whereas immunohistochemistry by a G1-specific antibody showed weak signals in vertebral tissues of Acan iE5/iE5 mice. Homozygous mutant mice are perinatally lethal and characterized by short snout, cleft palate and disproportionate dwarfism. Whole-mount skeletal staining and µ-CT analysis of Acan iE5/iE5 mice at embryonic day 18.5 revealed compressed vertebral bodies with accelerated mineralization compared to wild type controls. In Acan iE5/iE5 mice, histochemical staining revealed collapsed extracellular matrix with negligible sulfated glycosaminoglycan content accompanied by a high cellular density. Collagen type II deposition was not impaired in the IVD of Acan iE5/iE5 mice, as shown by immunohistochemistry. Mutant mice developed a severe IVD phenotype with deformed nucleus pulposus and thinned cartilaginous endplates accompanied by a disrupted growth plate structure in the vertebral body. Atomic force microscopy (AFM) imaging demonstrated a denser collagen network with thinner fibrils in the mutant IVD zones compared to wild type. Nanoscale AFM indentation revealed bimodal stiffness distribution attributable to the softer proteoglycan moiety and harder collagenous fibrils of the wild type IVD ECM. In Acan iE5/iE5 mice, loss of aggrecan resulted in a marked shift of the Young's modulus to higher values in all IVD zones. In conclusion, we demonstrated that aggrecan is pivotal for the determination and maintenance of the proper stiffness of IVD and vertebral tissues, which in turn could play an essential role in providing developmental biomechanical cues.

8.
J Mol Med (Berl) ; 99(6): 831-844, 2021 06.
Article in English | MEDLINE | ID: mdl-33609145

ABSTRACT

Glycogen synthase kinase (GSK) 3 acts to negatively regulate multiple signaling pathways, including canonical Wnt signaling. The two mammalian GSK3 proteins (alpha and beta) are at least partially redundant. While Gsk3a KO mice are viable and display a metabolic phenotype, abnormal neuronal development, and accelerated aging, Gsk3b KO animals die late in embryogenesis or at birth. Selective Gsk3b KO in bone delays development of some bones, whereas cartilage-specific Gsk3b KO mice are normal except for elevated levels of GSK3A protein. However, the collective role of these two GSK3 proteins in cartilage was not evaluated. To address this, we generated tamoxifen-inducible, cartilage-specific Gsk3a/Gsk3b KO (described as "cDKO") in juvenile mice and investigated their skeletal phenotypes. We found that cartilage-specific Gsk3a/Gsk3b deletion in young, skeletally immature mice causes precocious growth plate (GP) remodeling, culminating in shorter long bones and hence, growth retardation. These mice exhibit inefficient breathing patterns at later stages and fail to survive. The disrupted GP in cDKO mice showed progressive loss of cellular and proteoglycan components, and immunostaining for SOX9, while BGLAP (osteocalcin) and COL2A1 increased. In addition, we observed increased osteoclast recruitment and cell apoptosis. Surprisingly, changes in articular cartilage of cDKO mice were mild compared with the GP, signifying differential regulation of articular cartilage vs GP tissues. Taken together, these findings emphasize a crucial role of two GSK3 proteins in skeletal development, in particular in the maintenance and function of GP. KEY MESSAGES: • Both GSK3 genes, together, are crucial regulators of growth plate remodeling. • Cartilage-specific deletion of both GSK3 genes causes skeletal growth retardation. • Deletion of both GSK3 genes decreases Sox9 levels and promotes chondrocyte apoptosis. • Cartilage-specific GSK3 deletion in juvenile mice culminates in premature lethality. • GSK3 deletion exhibits mild effects on articular cartilage compared to growth plate.


Subject(s)
Gene Deletion , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3/genetics , Growth Plate/metabolism , Animals , Apoptosis/genetics , Biomarkers , Cartilage/metabolism , Chondrocytes/metabolism , Gene Knockdown Techniques , Mice , Mice, Knockout , Osteoclasts/metabolism
9.
Aging Cell ; 19(3): e13091, 2020 03.
Article in English | MEDLINE | ID: mdl-32083813

ABSTRACT

The intervertebral disc (IVD) degeneration is thought to be closely related to ingrowth of new blood vessels. However, the impact of anti-angiogenic factors in the maintenance of IVD avascularity remains unknown. Tenomodulin (Tnmd) is a tendon/ligament-specific marker and anti-angiogenic factor with abundant expression in the IVD. It is still unclear whether Tnmd contributes to the maintenance of IVD homeostasis, acting to inhibit vascular ingrowth into this normally avascular tissue. Herein, we investigated whether IVD degeneration could be induced spontaneously by the absence of Tnmd. Our results showed that Tnmd was expressed in an age-dependent manner primarily in the outer annulus fibrous (OAF) and it was downregulated at 6 months of age corresponding to the early IVD degeneration stage in mice. Tnmd knockout (Tnmd-/- ) mice exhibited more rapid progression of age-related IVD degeneration. These signs include smaller collagen fibril diameter, markedly lower compressive stiffness, reduced multiple IVD- and tendon/ligament-related gene expression, induced angiogenesis, and macrophage infiltration in OAF, as well as more hypertrophic-like chondrocytes in the nucleus pulposus. In addition, Tnmd and chondromodulin I (Chm1, the only homologous gene to Tnmd) double knockout (Tnmd-/- Chm1-/- ) mice displayed not only accelerated IVD degeneration, but also ectopic bone formation of IVD. Lastly, the absence of Tnmd in OAF-derived cells promoted p65 and matrix metalloproteinases upregulation, and increased migratory capacity of human umbilical vein endothelial cells. In sum, our data provide clear evidences that Tnmd acts as an angiogenic inhibitor in the IVD homeostasis and protects against age-related IVD degeneration. Targeting Tnmd may represent a novel therapeutic strategy for attenuating age-related IVD degeneration.


Subject(s)
Aging/metabolism , Disease Progression , Intervertebral Disc Degeneration/metabolism , Membrane Proteins/metabolism , Adult , Animals , Annulus Fibrosus/metabolism , Annulus Fibrosus/pathology , Cells, Cultured , Chondrocytes/metabolism , Coculture Techniques , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Gene Expression , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/pathology , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Nucleus Pulposus/metabolism , Nucleus Pulposus/pathology , Risk Factors , Young Adult
10.
Curr Top Dev Biol ; 133: 25-47, 2019.
Article in English | MEDLINE | ID: mdl-30902255

ABSTRACT

During cartilage development chondrocytes undergo a multi-step process characterized by consecutive changes in cell morphology and gene expression. Cell proliferation, polarity, differentiation, and migration are influenced by chemical and mechanical signaling between the extracellular matrix (ECM) and the cell. Several structurally diverse transmembrane receptors such as integrins, discoidin domain receptor 2 (DDR 2), and CD44 mediate the crosstalk between cells and their ECM. However, the contribution of cell-matrix interactions during early chondrogenesis and further cartilage development through cell receptors and their signal transduction pathways is still not fully understood. Determination of receptor signaling pathways and the function of downstream targets will aid in a better understanding of musculoskeletal pathologies such as chondrodysplasia, and the development of new approaches for the treatment of cartilage disorders. We will summarize recent findings, linking cell receptors and their potential signaling pathways to the control of chondrocyte behavior during early chondrogenesis and endochondral ossification.


Subject(s)
Cartilage/embryology , Cartilage/metabolism , Extracellular Matrix/metabolism , Osteogenesis , Signal Transduction , Animals , Chondrogenesis , Humans , Integrins/metabolism
11.
J Tissue Eng Regen Med ; 13(3): 359-368, 2019 03.
Article in English | MEDLINE | ID: mdl-30515986

ABSTRACT

Adipose-derived multipotent stem/progenitor cells (ASPCs) were shown to be ideal candidates for cell-based regenerative therapies. Yet, despite their huge potential, successful clinical applications are still rare. It was suggested that the efficacy of ASPCs at the recipient site depends on the vehicle of cell delivery. In this study, for preparation of a murine critical-size nerve defect model, we assessed the commercially available fibrin gel (ARTISS) as a potential cell carrier. In a thorough in vitro analysis, we investigated cell-fibrin interactions and analyzed the distribution and the long-term behavior of ASPCs cultivated in fibrin gel under normoxic and hypoxic conditions. ASPCs attached to the surface of a thin fibrin layer (two-dimensional condition) and spread with the abundant formation of actin stress fibers. Cells cultured within a fibrin matrix (three-dimensional condition) displayed a uniform distribution and formed interconnected networks while exhibiting strong cell-matrix interactions. Using time-lapse analysis, cells were found to migrate out of the gel and subsequently proliferated robustly both under hypoxic and normoxic conditions. During 14 days of culture in fibrin gel, ASPCs showed high viability, metabolic, and remodeling activities. At the end of the culture period, the fibrin matrix was degraded entirely accompanied by an upregulation of matrix metalloproteinases. In conclusion, fibrin gel stands out as a valuable biomaterial for delivering vital and active cells to damaged tissues. As a direct proof, ASPCs carried in a fibrin matrix will be evaluated in a murine critically sized peripheral nerve repair model.


Subject(s)
Adipose Tissue/cytology , Fibrin Tissue Adhesive/pharmacology , Regeneration/drug effects , Stem Cells/cytology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Biomechanical Phenomena , Cell Count , Cell Survival/drug effects , Fibrin/metabolism , Male , Matrix Metalloproteinases/metabolism , Mice , Rats, Wistar , Stem Cells/drug effects , Up-Regulation/drug effects
12.
Matrix Biol ; 50: 1-15, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26454027

ABSTRACT

The growth plate (GP) is a dynamic tissue driving bone elongation through chondrocyte proliferation, hypertrophy and matrix production. The extracellular matrix (ECM) is the major determinant of GP biomechanical properties and assumed to play a pivotal role for chondrocyte geometry and arrangement, thereby guiding proper growth plate morphogenesis and bone elongation. To elucidate the relationship between morphology and biomechanics during cartilage morphogenesis, we have investigated age-dependent structural and elastic properties of the proliferative zone of the murine GP by atomic force microscopy (AFM) from the embryonic stage to adulthood. We observed a progressive cell flattening and arrangement into columns from embryonic day 13.5 until postnatal week 2, correlating with an increasing collagen density and ECM stiffness, followed by a nearly constant cell shape, collagen density and ECM stiffness from week 2 to 4 months. At all ages, we found marked differences in the density and organization of the collagen network between the intracolumnar matrix, and the intercolumnar matrix, associated with a roughly two-fold higher stiffness of the intracolumnar matrix compared to the intercolumnar matrix. This difference in local ECM stiffness may force the cells to arrange in a columnar structure upon cell division and drive bone elongation during embryonic and juvenile development.


Subject(s)
Cartilage, Articular/growth & development , Growth Plate/physiology , Growth Plate/ultrastructure , Animals , Biomechanical Phenomena , Cartilage, Articular/physiology , Cell Proliferation , Extracellular Matrix/physiology , Growth Plate/growth & development , Mice , Microscopy, Atomic Force , Stress, Mechanical
13.
Matrix Biol ; 49: 132-143, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26429145

ABSTRACT

Collagen IX (Col IX) is an important component of the cartilage extracellularmatrix and has been associated with degenerative cartilage disorders and chondrodysplasias in humans. Further, polymorphisms in Col IX are known risk factors for the development of early intervertebral disc (IVD) degeneration. To understand the role of Col IX in the pathogenesis of IVD disorders, the spine of newborn and older Col IX deficient mice was systematically analyzed and compared to C57BL/6N controls. Morphology and bone parameters of the spine from newborn, 6 and 10 months old animals were investigated using µCT measurements. Histological staining was used to evaluate tissue structure and degree of degeneration. Localization and expression of extracellularmatrix proteins was analyzed in depth by immunofluorescence staining, immunoblotting, RT-PCR and in situ hybridization. High resolution imaging and stiffness measurements were performed by atomic force microscopy (AFM). Vertebral bodies of newborn Col IX-deficient mice were smaller and showed an increased mineral density compared to wild type animals. At birth, lack of Col IX led to a disrupted cellular organization in the cartilaginous endplate and a smaller nucleus pulposus of the IVD.Expression levels and localization of other extracellularmatrix proteins were strongly altered accompanied by a softening of cartilaginous tissues. In older animals, absence of Col IX caused earlier and more pronounced disc degeneration with annular fissures. The absence of Col IX induces early developmental, structural and biomechanical alterations in both vertebral body and intervertebral disc which eventually cause severe degenerative changes in the aging spine.


Subject(s)
Aging/pathology , Collagen Type IX/deficiency , Intervertebral Disc Degeneration/pathology , Spine/pathology , Aging/genetics , Aging/metabolism , Animals , Bone Density , Disease Models, Animal , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation , Humans , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Mice , Mice, Inbred C57BL
14.
Nat Commun ; 7: 13515, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27901020

ABSTRACT

Netrins, a family of laminin-related molecules, have been proposed to act as guidance cues either during nervous system development or the establishment of the vascular system. This was clearly demonstrated for netrin-1 via its interaction with the receptors DCC and UNC5s. However, mainly based on shared homologies with netrin-1, netrin-4 was also proposed to play a role in neuronal outgrowth and developmental/pathological angiogenesis via interactions with netrin-1 receptors. Here, we present the high-resolution structure of netrin-4, which shows unique features in comparison with netrin-1, and show that it does not bind directly to any of the known netrin-1 receptors. We show that netrin-4 disrupts laminin networks and basement membranes (BMs) through high-affinity binding to the laminin γ1 chain. We hypothesize that this laminin-related function is essential for the previously described effects on axon growth promotion and angiogenesis. Our study unveils netrin-4 as a non-enzymatic extracellular matrix protein actively disrupting pre-existing BMs.


Subject(s)
Axon Guidance/physiology , Basement Membrane/metabolism , Laminin/physiology , Neovascularization, Physiologic/physiology , Netrins/physiology , Animals , Axons/physiology , Chickens , Chorioallantoic Membrane/physiology , Crystallography, X-Ray , Female , HEK293 Cells , Humans , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Netrins/ultrastructure , Protein Binding , Protein Multimerization , Rats , Rats, Sprague-Dawley , Schwann Cells , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL