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










Publication year range
1.
EMBO Mol Med ; 15(7): e17528, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37292039

ABSTRACT

Osteogenesis imperfecta (OI) is a hereditary skeletal disorder primarily affecting collagen type I structure and function, causing bone fragility and occasionally versatile extraskeletal symptoms. This study expands the spectrum of OI-causing TAPT1 mutations and links extracellular matrix changes to signaling regulation.


Subject(s)
Osteogenesis Imperfecta , Humans , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/diagnosis , Collagen Type I/genetics , Extracellular Matrix , Mutation , Signal Transduction
2.
Am J Hum Genet ; 107(5): 989-999, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33053334

ABSTRACT

Osteogenesis imperfecta (OI) is characterized primarily by susceptibility to fractures with or without bone deformation. OI is genetically heterogeneous: over 20 genetic causes are recognized. We identified bi-allelic pathogenic KDELR2 variants as a cause of OI in four families. KDELR2 encodes KDEL endoplasmic reticulum protein retention receptor 2, which recycles ER-resident proteins with a KDEL-like peptide from the cis-Golgi to the ER through COPI retrograde transport. Analysis of patient primary fibroblasts showed intracellular decrease of HSP47 and FKBP65 along with reduced procollagen type I in culture media. Electron microscopy identified an abnormal quality of secreted collagen fibrils with increased amount of HSP47 bound to monomeric and multimeric collagen molecules. Mapping the identified KDELR2 variants onto the crystal structure of G. gallus KDELR2 indicated that these lead to an inactive receptor resulting in impaired KDELR2-mediated Golgi-ER transport. Therefore, in KDELR2-deficient individuals, OI most likely occurs because of the inability of HSP47 to bind KDELR2 and dissociate from collagen type I. Instead, HSP47 remains bound to collagen molecules extracellularly, disrupting fiber formation. This highlights the importance of intracellular recycling of ER-resident molecular chaperones for collagen type I and bone metabolism and a crucial role of HSP47 in the KDELR2-associated pathogenic mechanism leading to OI.


Subject(s)
Bone and Bones/metabolism , Collagen Type I/metabolism , HSP47 Heat-Shock Proteins/metabolism , Osteogenesis Imperfecta/genetics , Vesicular Transport Proteins/metabolism , Adult , Alleles , Amino Acid Sequence , Animals , Binding Sites , Bone and Bones/pathology , Chickens , Child, Preschool , Collagen Type I/chemistry , Collagen Type I/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , HSP47 Heat-Shock Proteins/chemistry , HSP47 Heat-Shock Proteins/genetics , Humans , Infant , Male , Osteogenesis Imperfecta/diagnosis , Osteogenesis Imperfecta/metabolism , Osteogenesis Imperfecta/pathology , Pedigree , Primary Cell Culture , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protein Transport , Sequence Alignment , Sequence Homology, Amino Acid , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics
3.
Cell Signal ; 76: 109789, 2020 12.
Article in English | MEDLINE | ID: mdl-32980496

ABSTRACT

Osteogenesis imperfecta (OI) is a clinically and genetically heterogeneous connective tissue disorder characterized by bone fragility and skeletal deformity. To maintain skeletal strength and integrity, bone undergoes constant remodeling of its extracellular matrix (ECM) tightly controlled by osteoclast-mediated bone resorption and osteoblast-mediated bone formation. There are at least 20 recognized OI-forms caused by mutations in the two collagen type I-encoding genes or genes implicated in collagen folding, posttranslational modifications or secretion of collagen, osteoblast differentiation and function, or bone mineralization. The underlying disease mechanisms of non-classical forms of OI that are not caused by collagen type I mutations are not yet completely understood, but an altered ECM structure as well as disturbed intracellular homeostasis seem to be the main defects. The ECM orchestrates local cell behavior in part by regulating bioavailability of signaling molecules through sequestration, release and activation during the constant bone remodeling process. Here, we provide an overview of signaling pathways that are associated with known OI-causing genes and discuss the impact of these genes on signal transduction. These pathways include WNT-, RANK/RANKL-, TGFß-, MAPK- and integrin-mediated signaling as well as the unfolded protein response.


Subject(s)
Osteogenesis Imperfecta/genetics , Transforming Growth Factor beta/genetics , Unfolded Protein Response/genetics , Wnt Signaling Pathway/genetics , Animals , Extracellular Matrix , Humans , Integrins/genetics , Mutation
4.
Mol Cell Pediatr ; 7(1): 9, 2020 Aug 14.
Article in English | MEDLINE | ID: mdl-32797291

ABSTRACT

Osteogenesis imperfecta (OI) is a rare congenital disease with a wide spectrum of severity characterized by skeletal deformity and increased bone fragility as well as additional, variable extraskeletal symptoms. Here, we present an overview of the genetic heterogeneity and pathophysiological background of OI as well as OI-related bone fragility disorders and highlight current therapeutic options.The most common form of OI is caused by mutations in the two collagen type I genes. Stop mutations usually lead to reduced collagen amount resulting in a mild phenotype, while missense mutations mainly provoke structural alterations in the collagen protein and entail a more severe phenotype. Numerous other causal genes have been identified during the last decade that are involved in collagen biosynthesis, modification and secretion, the differentiation and function of osteoblasts, and the maintenance of bone homeostasis.Management of patients with OI involves medical treatment by bisphosphonates as the most promising therapy to inhibit bone resorption and thereby facilitate bone formation. Surgical treatment ensures pain reduction and healing without an increase of deformities. Timely remobilization and regular strengthening of the muscles by physiotherapy are crucial to improve mobility, prevent muscle wasting and avoid bone resorption caused by immobilization. Identification of the pathomechanism for SERPINF1 mutations led to the development of a tailored mechanism-based therapy using denosumab, and unraveling further pathomechanisms will likely open new avenues for innovative treatment approaches.

5.
Int J Mol Sci ; 21(11)2020 Jun 09.
Article in English | MEDLINE | ID: mdl-32526967

ABSTRACT

MicroRNAs (miRNAs) regulate cartilage differentiation and contribute to the onset and progression of joint degeneration. These small RNA molecules may affect extracellular matrix organization (ECM) in cartilage, but for only a few miRNAs has this role been defined in vivo. Previously, we showed that cartilage-specific genetic ablation of the Mirc24 cluster in mice leads to impaired cartilage development due to increased RAF/MEK/ERK pathway activation. Here, we studied the expression of the cluster in cartilage by LacZ reporter gene assays and determined its role for extracellular matrix homeostasis by proteome and immunoblot analysis. The cluster is expressed in prehypertrophic/hypertrophic chondrocytes of the growth plate and we now show that the cluster is also highly expressed in articular cartilage. Cartilage-specific loss of the cluster leads to increased proteoglycan 4 and matrix metallopeptidase 13 levels and decreased aggrecan and collagen X levels in epiphyseal cartilage. Interestingly, these changes are linked to a decrease in SRY-related HMG box-containing (SOX) transcription factors 6 and 9, which regulate ECM production in chondrocytes. Our data suggests that the Mirc24 cluster is important for ECM homoeostasis and the expression of transcriptional regulators of matrix production in cartilage.


Subject(s)
Cartilage, Articular/metabolism , Extracellular Matrix Proteins/genetics , MicroRNAs/genetics , Osteoarthritis/genetics , Animals , Cartilage, Articular/physiology , Collagen Type II/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation , Growth Plate/chemistry , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Mice, Transgenic , Multigene Family , Proteoglycans/genetics , Proteoglycans/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , SOXD Transcription Factors/genetics , SOXD Transcription Factors/metabolism
6.
Int J Mol Sci ; 20(20)2019 Oct 14.
Article in English | MEDLINE | ID: mdl-31615030

ABSTRACT

The extracellular matrix (ECM) provides structural support for tissue architecture and is a major effector of cell behavior during skin repair and inflammation. Macrophages are involved in all stages of skin repair but only limited knowledge exists about macrophage-specific expression and regulation of ECM components. In this study, we used transcriptome profiling and bioinformatic analysis to define the unique expression of ECM-associated genes in cultured macrophages. Characterization of the matrisome revealed that most genes were constitutively expressed and that several genes were uniquely regulated upon interferon gamma (IFNγ) and dexamethasone stimulation. Among those core matrisome and matrisome-associated components transforming growth factor beta (TGFß)-induced, matrix metalloproteinase 9 (MMP9), elastin microfibril interfacer (EMILIN)-1, netrin-1 and gliomedin were also present within the wound bed at time points that are characterized by profound macrophage infiltration. Hence, macrophages are a source of ECM components in vitro as well as during skin wound healing, and identification of these matrisome components is a first step to understand the role and therapeutic value of ECM components in macrophages and during wound healing.


Subject(s)
Extracellular Matrix/genetics , Macrophages/metabolism , Skin/metabolism , Wound Healing/genetics , Animals , Computational Biology , Elastin/genetics , Gene Expression Profiling , Humans , Macrophages/pathology , Microarray Analysis , Skin/pathology
7.
J Cell Biol ; 218(6): 1853-1870, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31085560

ABSTRACT

In childhood, skeletal growth is driven by transient expansion of cartilage in the growth plate. The common belief is that energy production in this hypoxic tissue mainly relies on anaerobic glycolysis and not on mitochondrial respiratory chain (RC) activity. However, children with mitochondrial diseases causing RC dysfunction often present with short stature, which indicates that RC activity may be essential for cartilage-mediated skeletal growth. To elucidate the role of the mitochondrial RC in cartilage growth and pathology, we generated mice with impaired RC function in cartilage. These mice develop normally until birth, but their later growth is retarded. A detailed molecular analysis revealed that metabolic signaling and extracellular matrix formation is disturbed and induces cell death at the cartilage-bone junction to cause a chondrodysplasia-like phenotype. Hence, the results demonstrate the overall importance of the metabolic switch from fetal glycolysis to postnatal RC activation in growth plate cartilage and explain why RC dysfunction can cause short stature in children with mitochondrial diseases.


Subject(s)
Cartilage/pathology , Chondrocytes/pathology , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Growth Disorders/complications , Growth Plate/pathology , Mitochondrial Diseases/etiology , Animals , Cartilage/metabolism , Cell Differentiation , Chondrocytes/metabolism , Collagen Type II/physiology , DNA Helicases/physiology , Electron Transport , Energy Metabolism , Growth Disorders/metabolism , Growth Disorders/pathology , Growth Plate/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/physiology , Signal Transduction
8.
Stem Cells ; 36(11): 1752-1763, 2018 11.
Article in English | MEDLINE | ID: mdl-30063808

ABSTRACT

The trabecular extracellular matrix (ECM) forms a three-dimensional scaffold to stabilize the bone marrow, provide substrates for cell-matrix interactions and retain, present or release signals to modulate hematopoietic stem and progenitor cell development. However, the impact of trabecular ECM components on hematopoiesis has been poorly studied. Using collagen IX alpha1 - deficient (Col9a1(-/-) ) mice, we revealed that a lack of collagen IX alpha1 results in a disorganized trabecular network enriched in fibronectin, and in a reduction in myeloid cells, which was accompanied by a decrease in colony-stimulating factor 1 receptor expression on monocytes from the bone marrow. In contrast, B-cell numbers in the bone marrow and T-cell numbers in the thymus remained unchanged. Alterations in the bone marrow microenvironment may not only reduce myeloid cell numbers, but also have long-term implications for myeloid cell function. Mice were infected with Listeria moncytogenes to analyze the function of myeloid cells. In this case, an inadequate macrophage-dependent clearance of bacterial infections was observed in Col9a1(-/-) mice in vivo. This was mainly caused by an impaired interferon-gamma/tumor necrosis factor-alpha-mediated activation of macrophages. The loss of collagen IX alpha1 therefore destabilizes the trabecular bone network, impairs myeloid cell differentiation, and affects the innate immune response against Listeria. Stem Cells 2018;36:1752-1763.


Subject(s)
Collagen/metabolism , Myeloid Cells/metabolism , Animals , Disease Models, Animal , Female , Flow Cytometry , Humans , Mice
9.
Matrix Biol ; 65: 59-74, 2018 01.
Article in English | MEDLINE | ID: mdl-28797711

ABSTRACT

After skin injury fibroblasts migrate into the wound and transform into contractile, extracellular matrix-producing myofibroblasts to promote skin repair. Persistent activation of myofibroblasts can cause excessive fibrotic reactions, but the underlying mechanisms are not fully understood. We used SMA-GFP transgenic mice to study myofibroblast recruitment and activation in skin wounds. Myofibroblasts were initially recruited to wounds three days post injury, their number reached a maximum after seven days and subsequently declined. Expression profiling showed that 1749 genes were differentially expressed in sorted myofibroblasts from wounds seven days post injury. Most of these genes were linked with the extracellular region and cell periphery including genes encoding for extracellular matrix proteins. A unique panel of core matrisome and matrisome-associated genes was differentially expressed in myofibroblasts and several genes not yet known to be linked to myofibroblast-mediated wound healing were found (e.g. Col24a1, Podnl1, Bvcan, Tinagl1, Thbs3, Adamts16, Adamts19, Cxcl's, Ccl's). In addition, a complex network of G protein-coupled signaling events was regulated in myofibroblasts (e.g. Adcy1, Plbc4, Gnas). Hence, this first characterization of a myofibroblast-specific expression profile at the peak of in situ granulation tissue formation provides important insights into novel target genes that may control excessive ECM deposition during fibrotic reactions.


Subject(s)
Actins/genetics , Gene Expression Profiling/methods , Gene Regulatory Networks , Skin/injuries , Actins/metabolism , Animals , Cell Differentiation , Disease Models, Animal , Gene Expression Regulation , Mice , Mice, Transgenic , Myofibroblasts/chemistry , Myofibroblasts/cytology , Oligonucleotide Array Sequence Analysis , Organ Specificity , Skin/cytology , Skin/metabolism
10.
PLoS One ; 12(10): e0187030, 2017.
Article in English | MEDLINE | ID: mdl-29088242

ABSTRACT

Fetuin-A / α2-Heremans-Schmid-glycoprotein (gene name Ahsg) is a systemic inhibitor of ectopic calcification. Due to its high affinity for calcium phosphate, fetuin-A is highly abundant in mineralized bone matrix. Foreshortened femora in fetuin-A-deficient Ahsg-/- mice indicated a role for fetuin-A in bone formation. We studied early postnatal bone development in fetuin-A-deficient mice and discovered that femora from Ahsg-/- mice exhibited severely displaced distal epiphyses and deformed growth plates, similar to the human disease slipped capital femoral epiphysis (SCFE). The growth plate slippage occurred in 70% of Ahsg-/- mice of both sexes around three weeks postnatal. At this time point, mice weaned and rapidly gained weight and mobility. Epiphysis slippage never occurred in wildtype and heterozygous Ahsg+/- mice. Homozygous fetuin-A-deficient Ahsg-/- mice and, to a lesser degree, heterozygous Ahsg+/- mice showed lesions separating the proliferative zone from the hypertrophic zone of the growth plate. The hypertrophic growth plate cartilage in long bones from Ahsg-/- mice was significantly elongated and V-shaped until three weeks of age and thus prior to the slippage. Genome-wide transcriptome analysis of laser-dissected distal femoral growth plates from 13-day-old Ahsg-/- mice revealed a JAK-STAT-mediated inflammatory response including a 550-fold induction of the chemokine Cxcl9. At this stage, vascularization of the elongated growth plates was impaired, which was visualized by immunofluorescence staining. Thus, fetuin-A-deficient mice may serve as a rodent model of growth plate pathologies including SCFE and inflammatory cartilage degradation.


Subject(s)
Bone Diseases, Developmental/genetics , Epiphyses, Slipped/genetics , Femur/abnormalities , Hindlimb/abnormalities , alpha-2-HS-Glycoprotein/genetics , Animals , Female , Fluorescent Antibody Technique , Gene Expression Profiling/methods , Growth Plate/abnormalities , Male , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Weaning , alpha-2-HS-Glycoprotein/deficiency
11.
Development ; 144(19): 3562-3577, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28851708

ABSTRACT

Cartilage originates from mesenchymal cell condensations that differentiate into chondrocytes of transient growth plate cartilage or permanent cartilage of the articular joint surface and trachea. MicroRNAs fine-tune the activation of entire signaling networks and thereby modulate complex cellular responses, but so far only limited data are available on miRNAs that regulate cartilage development. Here, we characterize a miRNA that promotes the biosynthesis of a key component in the RAF/MEK/ERK pathway in cartilage. Specifically, by transcriptome profiling we identified miR-322 to be upregulated during chondrocyte differentiation. Among the various miR-322 target genes in the RAF/MEK/ERK pathway, only Mek1 was identified as a regulated target in chondrocytes. Surprisingly, an increased concentration of miR-322 stabilizes Mek1 mRNA to raise protein levels and dampen ERK1/2 phosphorylation, while cartilage-specific inactivation of miR322 in mice linked the loss of miR-322 to decreased MEK1 levels and to increased RAF/MEK/ERK pathway activation. Such mice died perinatally due to tracheal growth restriction and respiratory failure. Hence, a single miRNA can stimulate the production of an inhibitory component of a central signaling pathway to impair cartilage development.


Subject(s)
Cartilage/embryology , Cartilage/enzymology , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System , MicroRNAs/metabolism , Animals , Animals, Newborn , Binding Sites/genetics , CRISPR-Cas Systems/genetics , Chondrocytes/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Gene Silencing , Growth Plate/metabolism , Hemizygote , Homeostasis , MAP Kinase Kinase 1/genetics , Male , Mice, Transgenic , MicroRNAs/genetics , Organogenesis/genetics , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Transfection
12.
Connect Tissue Res ; 58(2): 196-207, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27386825

ABSTRACT

AIM: Wound healing is a coordinated process to restore tissue homeostasis and reestablish the protective barrier of the skin. miRNAs may modulate the expression of target genes to contribute to repair processes, but due to the complexity of the tissue it is challenging to quantify gene expression during the distinct phases of wound repair. Here, we aimed to identify a common reference gene to quantify changes in miRNA and mRNA expression during skin wound healing. METHODS: Quantitative real-time PCR and bioinformatic analysis tools were used to identify suitable reference genes during skin repair and their reliability was tested by studying the expression of mRNAs and miRNAs. RESULTS: Morphological assessment of wounds showed that the injury model recapitulates the distinct phases of skin repair. Non-degraded RNA could be isolated from skin and wounds and used to study the expression of non-coding small nuclear RNAs during wound healing. Among those, RNU6B was most constantly expressed during skin repair. Using this reference gene we could confirm the transient upregulation of IL-1ß and PTPRC/CD45 during the early phase as well as the increased expression of collagen type I at later stages of repair and validate the differential expression of miR-204, miR-205, and miR-31 in skin wounds. In contrast to Gapdh the normalization to multiple reference genes gave a similar outcome. CONCLUSION: RNU6B is an accurate alternative normalizer to quantify mRNA and miRNA expression during the distinct phases of skin wound healing when analysis of multiple reference genes is not feasible.


Subject(s)
Gene Expression Regulation , MicroRNAs/biosynthesis , RNA, Messenger/biosynthesis , Skin , Wound Healing , Wounds and Injuries/metabolism , Animals , Collagen Type I , Interleukin-1beta/biosynthesis , Leukocyte Common Antigens/biosynthesis , Mice , Skin/injuries , Skin/metabolism , Skin/pathology , Wounds and Injuries/pathology
13.
Cell Death Dis ; 7(11): e2488, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27882938

ABSTRACT

Systemic loss of neutral sphingomyelinase (SMPD3) in mice leads to a novel form of systemic, juvenile hypoplasia (dwarfism). SMPD3 deficiency in mainly two growth regulating cell types contributes to the phenotype, in chondrocytes of skeletal growth zones to skeletal malformation and chondrodysplasia, and in hypothalamic neurosecretory neurons to systemic hypothalamus-pituitary-somatotropic hypoplasia. The unbiased smpd3-/- mouse mutant and derived smpd3-/- primary chondrocytes were instrumental in defining the enigmatic role underlying the systemic and cell autonomous role of SMPD3 in the Golgi compartment. Here we describe the unprecedented role of SMPD3. SMPD3 deficiency disrupts homeostasis of sphingomyelin (SM), ceramide (Cer) and diacylglycerol (DAG) in the Golgi SMPD3-SMS1 (SM-synthase1) cycle. Cer and DAG, two fusogenic intermediates, modify the membrane lipid bilayer for the initiation of vesicle formation and transport. Dysproteostasis, unfolded protein response, endoplasmic reticulum stress and apoptosis perturb the Golgi secretory pathway in the smpd3-/- mouse. Secretion of extracellular matrix proteins is arrested in chondrocytes and causes skeletal malformation and chondrodysplasia. Similarly, retarded secretion of proteo-hormones in hypothalamic neurosecretory neurons leads to hypothalamus induced combined pituitary hormone deficiency. SMPD3 in the regulation of the protein vesicular secretory pathway may become a diagnostic target in the etiology of unknown forms of juvenile growth and developmental inhibition.


Subject(s)
Golgi Apparatus/metabolism , Secretory Pathway , Sphingomyelin Phosphodiesterase/deficiency , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Ceramides/metabolism , Chondrocytes/drug effects , Chondrocytes/metabolism , Detergents/pharmacology , Diglycerides/metabolism , Endoplasmic Reticulum Stress/drug effects , Gene Expression Profiling , Golgi Apparatus/drug effects , HEK293 Cells , Homeostasis/drug effects , Humans , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice, Inbred C57BL , Secretory Pathway/drug effects , Solubility , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism , Transferases (Other Substituted Phosphate Groups)/metabolism , Type C Phospholipases/metabolism , Unfolded Protein Response/drug effects
14.
Stem Cells ; 34(5): 1297-309, 2016 05.
Article in English | MEDLINE | ID: mdl-26934179

ABSTRACT

microRNAs (miRNAs) can regulate the interplay between perivascular cells (PVC) and endothelial cells (EC) during angiogenesis, but the relevant PVC-specific miRNAs are not yet defined. Here, we identified miR-126-3p and miR-146a to be exclusively upregulated in PVC upon interaction with EC, determined their influence on the PVC phenotype and elucidate their molecular mechanisms of action. Specifically the increase of miR-126-3p strongly promoted the motility of PVC on the basement membrane-like composite and stabilized networks of EC. Subsequent miRNA target analysis showed that miR-126-3p inhibits SPRED1 and PLK2 expression, induces ERK1/2 phosphorylation and stimulates TLR3 expression to modulate cell-cell and cell-matrix contacts of PVC. Gain of expression experiments in vivo demonstrated that miR-126-3p stimulates PVC coverage of newly formed vessels and transform immature into mature, less permeable vessels. In conclusion we showed that miR-126-3p regulates matrix-dependent PVC migration and intercellular interaction to modulate vascular integrity. Stem Cells 2016;34:1297-1309.


Subject(s)
Blood Vessels/cytology , Cell Communication/genetics , Cell Movement/genetics , Extracellular Matrix/metabolism , MicroRNAs/metabolism , Animals , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Communication/drug effects , Cell Movement/drug effects , Cell Shape/drug effects , Chemokines/metabolism , Coculture Techniques , Collagen/pharmacology , Drug Combinations , Extracellular Matrix/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Silencing/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Laminin/pharmacology , MAP Kinase Signaling System/drug effects , Mice , MicroRNAs/genetics , Neovascularization, Physiologic/genetics , Proteoglycans/pharmacology , Transcriptome/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
15.
Matrix Biol ; 43: 27-34, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25766405

ABSTRACT

MicroRNAs (miRNAs) may represent new therapeutic targets for bone and joint diseases. We hypothesized that several cartilage-specific proteins are targeted by a single miRNA and used bioinformatics to identify a miRNA that can modulate extracellular matrix (ECM) homeostasis in cartilage. Bioinformatic analysis of miRNA binding sequences in the 3'-untranslated region (3'-UTR) of target genes was performed to identify a miRNA that could bind to the 3'-UTR of cartilage matrix-related genes. MiRNA expression was studied by quantitative PCR of microdissected growth plate cartilage and binding to the 3'-UTR sequences was analyzed by luciferase interaction studies. Levels of proteins encoded by target genes in cultures of miR-26a mimic- or inhibitor-transfected chondrocytes were determined by FACS or immunoblot analysis. The complementary binding sequence of miR-26a and miR-26b was found in the 3'-UTR of the prehypertrophic/hypertrophic-specific genes Cd200, Col10a1 as well as Col9a1 and Ctgf. Both miRNAs were expressed in cartilage and only miR-26a was downregulated in hypertrophic growth plate cartilage. MiR-26a could interact with the 3'-UTR of Cd200 and Col10a1 in luciferase binding studies, but not with Col9a1 and Ctgf. However, protein expression of target genes and the ECM adaptor genes matrilin-3 and COMP was significantly altered in miR-26a mimic- or inhibitor-transfected chondrocytes, whereas the abundance of the cell surface receptor for insulin was not changed. In conclusion, miR-26a suppresses hypertrophic and ECM adaptor protein production. Dysregulation of miR-26a expression could contribute to ECM changes in cartilage diseases and this miRNA may therefore act as a therapeutic target.


Subject(s)
Cartilage Diseases/genetics , Chondrocytes/metabolism , Extracellular Matrix/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , 3' Untranslated Regions , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cartilage Diseases/pathology , Cells, Cultured , Chondrocytes/pathology , Collagen Type IX/genetics , Collagen Type IX/metabolism , Collagen Type X/genetics , Collagen Type X/metabolism , Computational Biology/methods , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Extracellular Matrix/genetics , Hypertrophy/genetics , Mice
16.
PLoS One ; 8(1): e53262, 2013.
Article in English | MEDLINE | ID: mdl-23308177

ABSTRACT

Skin injury induces the formation of new blood vessels by activating the vasculature in order to restore tissue homeostasis. Vascular cells may also differentiate into matrix-secreting contractile myofibroblasts to promote wound closure. Here, we characterize a PECAM1(+)/Sca1(+) vascular cell population in mouse skin, which is highly enriched in wounds at the peak of neoangiogenesis and myofibroblast formation. These cells express endothelial and perivascular markers and present the receptor CD38 on their surface. PECAM1(+)/Sca1(+)/CD38(+) cells proliferate upon wounding and could give rise to α-SMA(+) myofibroblast-like cells. CD38 stimulation in immunodeficient mice reduced the wound size at the peak of neoangiogenesis and myofibroblast formation. In humans a corresponding cell population was identified, which was enriched in sprouting vessels of basal cell carcinoma biopsies. The results indicate that PECAM1(+)/Sca1(+)/CD38(+) vascular cells could proliferate and differentiate into myofibroblast-like cells in wound repair. Moreover, CD38 signaling modulates PECAM1(+)/Sca1(+)/CD38(+) cell activation in the healing process implying CD38 as a target for anti-angiogenic therapies in human basal cell carcinoma.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Antigens, Ly/metabolism , Membrane Proteins/metabolism , Myofibroblasts/cytology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Skin Physiological Phenomena , Skin/blood supply , Wound Healing , ADP-ribosyl Cyclase 1/analysis , Animals , Antigens, Ly/analysis , Carcinoma, Basal Cell/metabolism , Foreskin/metabolism , Foreskin/ultrastructure , Humans , Male , Membrane Proteins/analysis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myofibroblasts/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/analysis
17.
J Biol Chem ; 286(7): 5708-16, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21131363

ABSTRACT

Identification and clearance of apoptotic cells prevents the release of harmful cell contents thereby suppressing inflammation and autoimmune reactions. Highly conserved annexins may modulate the phagocytic cell removal by acting as bridging molecules to phosphatidylserine, a characteristic phagocytosis signal of dying cells. In this study five members of the structurally and functionally related annexin family were characterized for their capacity to interact with phosphatidylserine and dying cells. The results showed that AnxA3, AnxA4, AnxA13, and the already described interaction partner AnxA5 can bind to phosphatidylserine and apoptotic cells, whereas AnxA8 lacks this ability. Sequence alignment experiments located the essential amino residues for the recognition of surface exposed phosphatidylserine within the calcium binding motifs common to all annexins. These amino acid residues were missing in the evolutionary young AnxA8 and when they were reintroduced by site directed mutagenesis AnxA8 gains the capability to interact with phosphatidylserine containing liposomes and apoptotic cells. By defining the evolutionary conserved amino acid residues mediating phosphatidylserine binding of annexins we show that the recognition of dying cells represent a common feature of most annexins. Hence, the individual annexin repertoire bound to the cell surface of dying cells may fulfil opsonin-like function in cell death recognition.


Subject(s)
Annexins/metabolism , Apoptosis/physiology , Calcium/metabolism , Phosphatidylserines/metabolism , Signal Transduction/physiology , Amino Acid Motifs , Animals , Annexins/chemistry , Annexins/genetics , Evolution, Molecular , Mice , NIH 3T3 Cells , Phosphatidylserines/chemistry , Phosphatidylserines/genetics , Protein Binding
18.
J Bone Miner Res ; 25(6): 1267-81, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20200945

ABSTRACT

Axial growth of long bones occurs through a coordinated process of growth plate chondrocyte proliferation and differentiation. This maturation of chondrocytes is reflected in a zonal change in gene expression and cell morphology from resting to proliferative, prehypertrophic, and hypertrophic chondrocytes of the growth plate followed by ossification. A major experimental limitation in understanding growth plate biology and pathophysiology is the lack of a robust technique to isolate cells from the different zones, particularly from small animals. Here, we report on a new strategy for separating distinct chondrocyte populations from mouse growth plates. By transcriptome profiling of microdissected zones of growth plates, we identified novel, zone-specific cell surface markers and used these for flow cytometry and immunomagnetic cell separation to quantify, enrich, and characterize chondrocytes populations with respect to their differentiation status. This approach provides a novel platform to study cartilage development and characterize mouse growth plate chondrocytes to reveal unique cellular phenotypes of the distinct subpopulations within the growth plate.


Subject(s)
Cell Differentiation , Cell Separation/methods , Chondrocytes/cytology , Flow Cytometry/methods , Growth Plate/cytology , Animals , Antigens, Surface/metabolism , Biomarkers/metabolism , Cell Death , Cell Membrane/metabolism , Chondrocytes/metabolism , Growth Plate/growth & development , Immunomagnetic Separation , Mice , Mice, Inbred C57BL , Microspheres , Organ Specificity , Phenotype , Reproducibility of Results
19.
J Bone Miner Res ; 25(1): 141-53, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19580468

ABSTRACT

Initiation of mineralization during endochondral ossification is a multistep process and has been assumed to correlate with specific interactions of annexins A5 and A6 and collagens. However, skeletal development appears to be normal in mice deficient for either A5 or A6, and the highly conserved structures led to the assumption that A5 and A6 may fulfill redundant functions. We have now generated mice deficient of both proteins. These mice were viable and fertile and showed no obvious abnormalities. Assessment of skeletal elements using histologic, ultrastructural, and peripheral quantitative computed tomographic methods revealed that mineralization and development of the skeleton were not significantly affected in mutant mice. Otherwise, global gene expression analysis showed subtle changes at the transcriptome level of genes involved in cell growth and intermediate metabolism. These results indicate that annexins A5 and A6 may not represent the essential annexins that promote mineralization in vivo.


Subject(s)
Annexin A5/deficiency , Annexin A6/deficiency , Calcification, Physiologic/genetics , Cartilage/metabolism , Gene Expression Profiling , Growth Plate/metabolism , Animals , Animals, Newborn , Annexin A5/genetics , Annexin A5/metabolism , Annexin A6/genetics , Annexin A6/metabolism , Antibody Specificity , Bone Development/genetics , Bone Matrix/metabolism , Bone Matrix/ultrastructure , Cartilage/ultrastructure , Cell Proliferation , Collagen/metabolism , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , Femur/growth & development , Femur/metabolism , Femur/ultrastructure , Gene Expression Regulation, Developmental , Growth Plate/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
20.
Exp Cell Res ; 313(12): 2730-43, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17543301

ABSTRACT

Pericytes are closely associated with endothelial cells, contribute to vascular stability and represent a potential source of mesenchymal progenitor cells. Using the specifically expressed annexin A5-LacZ fusion gene (Anxa5-LacZ), it became possible to isolate perivascular cells (PVC) from mouse tissues. These cells proliferate and can be cultured without undergoing senescence for multiple passages. PVC display phenotypic characteristics of pericytes, as they express pericyte-specific markers (NG2-proteoglycan, desmin, alphaSMA, PDGFR-beta). They also express stem cell marker Sca-1, whereas endothelial (PECAM), hematopoietic (CD45) or myeloid (F4/80, CD11b) lineage markers are not detectable. These characteristics are in common with the pericyte-like cell line 10T1/2. PVC also display a phagocytoic activity higher than 10T1/2 cells. During coculture with endothelial cells both cell types stimulate angiogenic processes indicated by an increased expression of PECAM in endothelial cells and specific deposition of basement membrane proteins. PVC show a significantly increased induction of endothelial specific PECAM expression compared to 10T1/2 cells. Accordingly, in vivo grafts of PVC aggregates onto chorioallantoic membranes of quail embryos recruit endothelial cells, get highly vascularized and deposit basement membrane components. These data demonstrate that isolated Anxa5-LacZ(+) PVC from mouse meninges retain their capacity for differentiation to pericyte-like cells and contribute to angiogenic processes.


Subject(s)
Annexin A5/metabolism , Meninges/blood supply , Meninges/cytology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Pericytes/cytology , Animals , Ataxin-1 , Ataxins , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Cell Separation , Cells, Cultured , Cellular Senescence , Coculture Techniques , Endothelial Cells/cytology , Humans , Mice , Neovascularization, Physiologic , Phagocytosis , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...