Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Arthritis Rheumatol ; 75(7): 1152-1165, 2023 07.
Article in English | MEDLINE | ID: mdl-36657110

ABSTRACT

OBJECTIVE: Spondyloarthritis (SpA) is characterized by pathologic osteogenesis, inflammation, and extensive angiogenesis in axial and peripheral tissues. Current therapies effectively target inflammation, but these therapies lack efficacy in preventing pathologic osteogenesis. Transgenic mice overexpressing transmembrane tumor necrosis factor (tmTNF-Tg mice) exhibit SpA-like features. We hypothesized that type H blood vessels, which are implicated in osteogenesis, are increased and contribute to pathology in this experimental SpA model. METHODS: We analyzed ankles, femora, and vertebrae of tmTNF-Tg mice and nontransgenic littermates and tmTNF-Tg mice on either a TNF receptor type I (TNFRI)-deficient or TNF receptor type II (TNFRII)-deficient background for osteogenesis, angiogenesis, and inflammation using advanced imaging technologies at various stages of disease. RESULTS: Compared to nontransgenic littermates, tmTNF-Tg mice exhibited an increase in vertebral type H vessels and osteoprogenitor cells in subchondral bone. These features of increased angiogenesis and osteogenesis were already present before onset of clinical disease symptoms. Type H vessels and osteoprogenitor cells were in close proximity to inflammatory lesions and ectopic lymphoid structures. The tmTNF-Tg mice also showed perivertebral ectopic type H vessels and osteogenesis, an increased number of vertebral transcortical vessels, and enhanced entheseal angiogenesis. In tmTNF-Tg mice crossed on a TNFRI- or TNFRII-deficient background, no clear reduction in type H vessels was shown, suggesting that type H vessel formation is not exclusively mediated via TNFRI or TNFRII. CONCLUSION: The contribution of type H vessels to pathologic osteogenesis in experimental SpA advances our knowledge of the pathophysiology of this disease and may also provide a novel opportunity for targeted intervention.


Subject(s)
Osteogenesis , Spondylarthritis , Mice , Animals , Inflammation , Spondylarthritis/drug therapy , Mice, Transgenic , Tumor Necrosis Factor-alpha
2.
Exp Dermatol ; 31(9): 1431-1442, 2022 09.
Article in English | MEDLINE | ID: mdl-35620886

ABSTRACT

Recessive dystrophic epidermolysis bullosa (RDEB) patients develop poorly healing skin wounds that are frequently colonized with microbiota. Because T cells play an important role in clearing such pathogens, we aimed to define the status of adaptive T cell-mediated immunity in RDEB wounds. Using a non-invasive approach for sampling of wound-associated constituents, we evaluated microbial contaminants in cellular fraction and exudates obtained from RDED wounds. Infectivity and intracellular trafficking of inactivated Staphylococcus aureus was accessed in RDEB keratinocytes. S. aureus and microbial antigen-specific activation of RDEB wound-derived T cells were investigated by fluorescence-activated cell sorting-based immune-phenotyping and T-cell functional assays. We found that RDEB wounds and epithelial cells are most frequently infected with Staphylococcus sp. and Pseudomonas sp. and that S. aureus essentially infects more RDEB keratinocytes and RDEB-derived squamous cell carcinoma cells than keratinocytes from healthy donors. The RDEB wound-associated T cells contain populations of CD4+ and CD8+ peripheral memory T cells that respond to soluble microbial antigens by proliferating and secreting interferon gamma (IFNγ). Moreover, CD8+ cytotoxic T lymphocytes recognize S. aureus-infected RDEB keratinocytes and respond by producing interleukin-2 (IL-2) and IFNγ and degranulating and cytotoxically killing infected cells. Prolonged exposure of RDEB-derived T cells to microbial antigens in vitro does not trigger PD-1-mediated T-cell exhaustion but induces differentiation of the CD4high population into CD4high CD25+ FoxP3+ regulatory T cells. Our data demonstrated that adaptive T cell-mediated immunity could clear infected cells from wound sites, but these effects might be inhibited by PD-1/Treg-mediated immuno-suppression in RDEB.


Subject(s)
Bacterial Infections , Epidermolysis Bullosa Dystrophica , T-Lymphocytes , Antigens , Collagen Type VII , Epidermolysis Bullosa Dystrophica/pathology , Humans , Keratinocytes/pathology , Lymphocyte Activation , Programmed Cell Death 1 Receptor , Staphylococcus aureus , T-Lymphocytes/immunology
3.
Exp Dermatol ; 30(12): 1724-1733, 2021 12.
Article in English | MEDLINE | ID: mdl-34142388

ABSTRACT

Hereditary epidermolysis bullosa (EB) is a mechanobullous skin fragility disorder characterized by defective epithelial adhesion, leading to mechanical stress-induced skin blistering. Based on the level of tissue separation within the dermal-epidermal junction, EB is categorized into simplex (EBS), junctional (JEB), dystrophic (DEB) and Kindler syndrome. There is no cure for EB, and painful chronic cutaneous wounds are one of the major complications in recessive (RDEB) patients. Although RDEB is considered a cutaneous disease, recent data support the underlying systemic immunological defects. Furthermore, chronic wounds are often colonized with pathogenic microbiota, leading to excessive inflammation and altered wound healing. Consequently, patients with RDEB suffer from a painful sensation of chronic, cutaneous itching/burning and an endless battle with bacterial infections. To improve their quality of life and life expectancy, it is important to prevent cutaneous infections, dampen chronic inflammation and stimulate wound healing. A clear scientific understanding of the immunological events underlying the maintenance of chronic poorly healing wounds in RDEB patients is necessary to improve disease management and better understand other wound healing disorders. In this review, we summarize current knowledge of the role of professional phagocytes, such as neutrophils, macrophages and dendritic cells, the role of T-cell-mediated immunity in lymphoid organs, and the association of microbiota with poor wound healing in RDEB. We conclude that RDEB patients have an underlying immunity defect that seems to affect antibacterial immunity.


Subject(s)
Epidermolysis Bullosa Dystrophica/physiopathology , Skin/pathology , Wound Healing , Epidermolysis Bullosa Dystrophica/immunology , Humans
4.
Exp Dermatol ; 30(10): 1428-1439, 2021 10.
Article in English | MEDLINE | ID: mdl-33179358

ABSTRACT

Pathogenic invasion of Staphylococcus aureus is a major concern in patients with chronic skin diseases like atopic dermatitis (AD), epidermolysis bullosa (EB), or chronic diabetic foot and venous leg ulcers, and can result in persistent and life-threatening chronic non-healing wounds. Staphylococcus aureus is generally recognized as extracellular pathogens. However, S. aureus can also invade, hide and persist in skin cells to contribute to wound chronicity. The intracellular life cycle of S. aureus is currently incompletely understood, although published studies indicate that its intracellular escape strategies play an important role in persistent cutaneous infections. This review provides current scientific knowledge about the intracellular life cycle of S. aureus in skin cells, which can be classified into professional and non-professional antigen-presenting cells, and its strategies to escape adaptive defense mechanisms. First, we discuss phenotypic switch of S. aureus, which affects intracellular routing and degradation. This review also evaluates potential intracellular escape mechanism of S. aureus to avoid intracellular degradation and antigen presentation, preventing an immune response. Furthermore, we discuss potential drug targets that can interfere with the intracellular life cycle of S. aureus. Taken together, this review aimed to increase scientific understanding about the intracellular life cycle of S. aureus into skin cells and its strategies to evade the host immune response, information that is crucial to reduce pathogenic invasion and life-threatening persistence of S. aureus in chronic cutaneous infections.


Subject(s)
Skin Diseases/immunology , Skin Diseases/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Autophagy , Humans , Staphylococcus aureus
5.
J Dermatol Sci ; 100(3): 209-216, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33143962

ABSTRACT

BACKGROUND: Poorly healing wounds are one of the major complications in patients suffering from recessive dystrophic epidermolysis bullosa (RDEB). At present, there are no effective means to analyze changes in cellular and molecular networks occurring during RDEB wound progression to predict wound outcome and design betted wound management approaches. OBJECTIVES: To better define mechanisms influencing RDEB wound progression by evaluating changes in molecular and cellular networks. METHODS: We developed a non-invasive approach for sampling and analysis of wound-associated constituents using wound-covering bandages. Cellular and molecular components from seventy-six samples collected from early, established and chronic RDEB wounds were evaluated by FACS-based immuno-phenotyping and ELISA. RESULTS: Our cross-sectional analysis determined that progression of RDEB wounds to chronic state is associated with the accumulation (up to 90 %) of CD16+CD66b+ mature neutrophils, loss of CD11b+CD68+ macrophages, and a significant increase (up to 50 %) in a number of CD11c+CD80+CD86+ activated professional antigen presenting cells (APC). It was also marked by changes in activated T cells populations including a reduction of CD45RO+ peripheral memory T cells from 80 % to 30 % and an increase (up to 70 %) in CD45RA+ effector T cells. Significantly higher levels of MMP9, VEGF-A and cathepsin G were also associated with advancing of wounds to poorly healing state. CONCLUSIONS: Our data demonstrated that wound-covering bandages are useful for a non-invasive sampling and analysis of wound-associated constituents and that transition to poorly healing wounds in RDEB patients as associated with distinct changes in leukocytic infiltrates, matrix-remodeling enzymes and pro-angiogenic factors at wound sites.


Subject(s)
Epidermolysis Bullosa Dystrophica/complications , Leukocytes/immunology , Skin/pathology , Wound Healing/immunology , Adolescent , Adult , Aged , Child , Child, Preschool , Cross-Sectional Studies , Epidermolysis Bullosa Dystrophica/immunology , Epidermolysis Bullosa Dystrophica/pathology , Female , Humans , Infant , Leukocytes/metabolism , Male , Middle Aged , Receptors, CCR2/metabolism , Receptors, Interleukin-8B/metabolism , Skin/cytology , Skin/immunology , Young Adult
6.
Elife ; 72018 04 06.
Article in English | MEDLINE | ID: mdl-29624170

ABSTRACT

Segmentation of the axial skeleton in amniotes depends on the segmentation clock, which patterns the paraxial mesoderm and the sclerotome. While the segmentation clock clearly operates in teleosts, the role of the sclerotome in establishing the axial skeleton is unclear. We severely disrupt zebrafish paraxial segmentation, yet observe a largely normal segmentation process of the chordacentra. We demonstrate that axial entpd5+ notochord sheath cells are responsible for chordacentrum mineralization, and serve as a marker for axial segmentation. While autonomous within the notochord sheath, entpd5 expression and centrum formation show some plasticity and can respond to myotome pattern. These observations reveal for the first time the dynamics of notochord segmentation in a teleost, and are consistent with an autonomous patterning mechanism that is influenced, but not determined by adjacent paraxial mesoderm. This behavior is not consistent with a clock-type mechanism in the notochord.


Subject(s)
Animals, Genetically Modified/physiology , Biological Clocks , Body Patterning , Bone and Bones/physiology , Notochord/physiology , Pyrophosphatases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/physiology , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Bone and Bones/embryology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/physiology , Gene Expression Regulation, Developmental , Mesoderm/embryology , Mesoderm/physiology , Mutation , Notochord/embryology , Pyrophosphatases/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
7.
Cell Rep ; 22(8): 2026-2038, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466731

ABSTRACT

The spine is a segmented axial structure made of alternating vertebral bodies (centra) and intervertebral discs (IVDs) assembled around the notochord. Here, we show that, prior to centra formation, the outer epithelial cell layer of the zebrafish notochord, the sheath, segments into alternating domains corresponding to the prospective centra and IVD areas. This process occurs sequentially in an anteroposterior direction via the activation of Notch signaling in alternating segments of the sheath, which transition from cartilaginous to mineralizing domains. Subsequently, osteoblasts are recruited to the mineralized domains of the notochord sheath to form mature centra. Tissue-specific manipulation of Notch signaling in sheath cells produces notochord segmentation defects that are mirrored in the spine. Together, our findings demonstrate that notochord sheath segmentation provides a template for vertebral patterning in the zebrafish spine.


Subject(s)
Body Patterning , Notochord/embryology , Spine/embryology , Zebrafish/embryology , Animals , Cartilage/metabolism , Gene Expression Regulation, Developmental , Morphogenesis , Osteoblasts/metabolism , Receptors, Notch/metabolism , Signal Transduction , Somites/metabolism
8.
Elife ; 72018 02 06.
Article in English | MEDLINE | ID: mdl-29405914

ABSTRACT

Regenerative therapy for degenerative spine disorders requires the identification of cells that can slow down and possibly reverse degenerative processes. Here, we identify an unanticipated wound-specific notochord sheath cell subpopulation that expresses Wilms Tumor (WT) 1b following injury in zebrafish. We show that localized damage leads to Wt1b expression in sheath cells, and that wt1b+cells migrate into the wound to form a stopper-like structure, likely to maintain structural integrity. Wt1b+sheath cells are distinct in expressing cartilage and vacuolar genes, and in repressing a Wt1b-p53 transcriptional programme. At the wound, wt1b+and entpd5+ cells constitute separate, tightly-associated subpopulations. Surprisingly, wt1b expression at the site of injury is maintained even into adult stages in developing vertebrae, which form in an untypical manner via a cartilage intermediate. Given that notochord cells are retained in adult intervertebral discs, the identification of novel subpopulations may have important implications for regenerative spine disorder treatments.


Subject(s)
Nerve Regeneration , Neuroglia/chemistry , Neuroglia/physiology , Notochord/injuries , WT1 Proteins/analysis , Wound Healing , Animals , Cell Movement , Zebrafish
9.
Front Immunol ; 9: 2902, 2018.
Article in English | MEDLINE | ID: mdl-30692988

ABSTRACT

Expression of the Autoimmune regulator (AIRE) outside of the thymus has long been suggested in both humans and mice, but the cellular source in humans has remained undefined. Here we identify AIRE expression in human tonsils and extensively analyzed these "extra-thymic AIRE expressing cells" (eTACs) using combinations of flow cytometry, CyTOF and single cell RNA-sequencing. We identified AIRE+ cells as dendritic cells (DCs) with a mature and migratory phenotype including high levels of antigen presenting molecules and costimulatory molecules, and specific expression of CD127, CCR7, and PDL1. These cells also possessed the ability to stimulate and re-stimulate T cells and displayed reduced responses to toll-like receptor (TLR) agonists compared to conventional DCs. While expression of AIRE was enriched within CCR7+CD127+ DCs, single-cell RNA sequencing revealed expression of AIRE to be transient, rather than stable, and associated with the differentiation to a mature phenotype. The role of AIRE in central tolerance induction within the thymus is well-established, however our study shows that AIRE expression within the periphery is not associated with an enriched expression of tissue-restricted antigens (TRAs). This unexpected finding, suggestive of wider functions of AIRE, may provide an explanation for the non-autoimmune symptoms of APECED patients who lack functional AIRE.


Subject(s)
Dendritic Cells/immunology , Palatine Tonsil/cytology , Transcription Factors/metabolism , B7-H1 Antigen/metabolism , Cell Communication/immunology , Cells, Cultured , Child, Preschool , Dendritic Cells/metabolism , Humans , Infant , Infant, Newborn , Interleukin-7 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Palatine Tonsil/immunology , Palatine Tonsil/metabolism , Primary Cell Culture , Receptors, CCR7/metabolism , T-Lymphocytes/immunology , Transcription Factors/immunology , AIRE Protein
10.
Cell Metab ; 25(1): 197-207, 2017 01 10.
Article in English | MEDLINE | ID: mdl-27866837

ABSTRACT

Membrane cholesterol modulates a variety of cell signaling pathways and functions. While cholesterol depletion by high-density lipoproteins (HDLs) has potent anti-inflammatory effects in various cell types, its effects on inflammatory responses in macrophages remain elusive. Here we show overt pro-inflammatory effects of HDL-mediated passive cholesterol depletion and lipid raft disruption in murine and human primary macrophages in vitro. These pro-inflammatory effects were confirmed in vivo in peritoneal macrophages from apoA-I transgenic mice, which have elevated HDL levels. In line with these findings, the innate immune responses required for clearance of P. aeruginosa bacterial infection in lung were compromised in mice with low HDL levels. Expression analysis, ChIP-PCR, and combinatorial pharmacological and genetic intervention studies unveiled that both native and reconstituted HDL enhance Toll-like-receptor-induced signaling by activating a PKC-NF-κB/STAT1-IRF1 axis, leading to increased inflammatory cytokine expression. HDL's pro-inflammatory activity supports proper functioning of macrophage immune responses.


Subject(s)
Cholesterol/metabolism , Inflammation/metabolism , Inflammation/pathology , Lipoproteins, HDL/pharmacology , Macrophages/metabolism , Macrophages/pathology , Signal Transduction/drug effects , Animals , Base Sequence , Biological Transport/drug effects , Cells, Cultured , Humans , Interferon Regulatory Factor-1/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , NF-kappa B/metabolism , Protein Kinase C/metabolism , Respiratory Tract Infections/metabolism , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/pathology , STAT1 Transcription Factor/metabolism , Toll-Like Receptors/metabolism
11.
Eur J Clin Invest ; 45(5): 529-39, 2015 May.
Article in English | MEDLINE | ID: mdl-25735405

ABSTRACT

BACKGROUND: Immune regulation is necessary to control inflammatory responses and to prevent autoimmune diseases. Therefore, mechanisms of central and peripheral tolerance have evolved to ensure that T cells recognize antigens as self- or non-self-antigens. The thymus is crucially important for central tolerance induction to self-antigens via negative selection of T cells. However, if T cells escape negative selection in the thymus and enter the periphery, peripheral mechanisms are active to warrant immune tolerance. Secondary lymphoid organs, as well as tolerogenic dendritic cells and regulatory T cells, play an important role in peripheral tolerance. In chronic inflammatory diseases, tertiary lymphoid organs are sometimes formed that may also be involved in the induction of peripheral tolerance. This review discusses the main processes that are involved in immune regulation and tolerance, and focuses on the contribution of NF-κB signalling to these processes. MATERIAL AND METHODS: This narrative review is based on peer-reviewed publications listed on PubMed up to December 2014. The focus of our literature search was on studies investigating the role of (non)canonical NF-κB signalling in central and peripheral mechanisms of tolerance. Only studies published in English language were considered. RESULTS: This review discusses the immune phenotype of mutant mice with defective (non)canonical NF-κB signalling, corroborated with human data, and emphasizes the contribution of the noncanonical NF-κB pathway to immune regulation and tolerance induction. CONCLUSIONS: Noncanonical NF-κB signalling has an important immunoregulatory role in the immune system and contributes to both central and peripheral mechanisms of tolerance.


Subject(s)
Immune Tolerance/immunology , NF-kappa B/immunology , T-Lymphocytes/immunology , Animals , Autoimmune Diseases/immunology , Dendritic Cells/immunology , Humans , Lymphoid Tissue/immunology , Mice , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology
12.
Zebrafish ; 11(6): 542-50, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25353089

ABSTRACT

Several genome-wide association studies have identified the genes encoding for macrophage-stimulating protein (MSP) and its receptor RON (Recepteur d'Origine Nantais) as possible susceptibility factors in inflammatory bowel disease. While it has been shown that the MSP-RON signaling pathway is involved in tissue injury responses, current mouse models for MSP and RON deficiency have not clearly demonstrated a role of MSP-RON signaling in the context of intestinal inflammation. In this study, we report that the recently identified zebrafish Msp mutant (msp(t34230)) develops spontaneous intestinal inflammation over time. From 14 to 28 weeks postfertilization Msp-deficient zebrafish show intestinal eosinophilia, increased intestinal expression of inflammatory marker mmp9, and activation of intestinal goblet cells. Moreover, these Msp mutant zebrafish are more susceptible toward ethanol-induced epithelial damage, which resulted in increased infiltration and proliferation of immune cells within the lamina propria and prolonged intestinal proinflammatory cytokine responses in some mutant fish. In light of the recent development of many tools to visualize, monitor, and genetically modify zebrafish, these Msp-deficient zebrafish will enable in-depth in vivo analysis of epithelial and macrophage-specific MSP-RON signaling in the context of intestinal inflammation.


Subject(s)
Disease Models, Animal , Hepatocyte Growth Factor/deficiency , Inflammation/genetics , Inflammation/pathology , Intestinal Mucosa/metabolism , Proto-Oncogene Proteins/deficiency , Signal Transduction/genetics , Zebrafish , Animals , Cytokines , DNA Primers/genetics , Goblet Cells/metabolism , Hepatocyte Growth Factor/genetics , Histological Techniques , Immunohistochemistry , Intestinal Mucosa/pathology , Matrix Metalloproteinase 9/metabolism , Mutation/genetics , Proto-Oncogene Proteins/genetics , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric
13.
Dis Model Mech ; 7(7): 811-22, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24906371

ABSTRACT

In recent years it has become clear that, mechanistically, biomineralization is a process that has to be actively inhibited as a default state. This inhibition must be released in a rigidly controlled manner in order for mineralization to occur in skeletal elements and teeth. A central aspect of this concept is the tightly controlled balance between phosphate, a constituent of the biomineral hydroxyapatite, and pyrophosphate, a physiochemical inhibitor of mineralization. Here, we provide a detailed analysis of a zebrafish mutant, dragonfish (dgf), which is mutant for ectonucleoside pyrophosphatase/phosphodiesterase 1 (Enpp1), a protein that is crucial for supplying extracellular pyrophosphate. Generalized arterial calcification of infancy (GACI) is a fatal human disease, and the majority of cases are thought to be caused by mutations in ENPP1. Furthermore, some cases of pseudoxanthoma elasticum (PXE) have recently been linked to ENPP1. Similar to humans, we show here that zebrafish enpp1 mutants can develop ectopic calcifications in a variety of soft tissues - most notably in the skin, cartilage elements, the heart, intracranial space and the notochord sheet. Using transgenic reporter lines, we demonstrate that ectopic mineralizations in these tissues occur independently of the expression of typical osteoblast or cartilage markers. Intriguingly, we detect cells expressing the osteoclast markers Trap and CathepsinK at sites of ectopic calcification at time points when osteoclasts are not yet present in wild-type siblings. Treatment with the bisphosphonate etidronate rescues aspects of the dgf phenotype, and we detected deregulated expression of genes that are involved in phosphate homeostasis and mineralization, such as fgf23, npt2a, entpd5 and spp1 (also known as osteopontin). Employing a UAS-GalFF approach, we show that forced expression of enpp1 in blood vessels or the floorplate of mutant embryos is sufficient to rescue the notochord mineralization phenotype. This indicates that enpp1 can exert its function in tissues that are remote from its site of expression.


Subject(s)
Calcinosis/complications , Mutation/genetics , Phosphoric Diester Hydrolases/genetics , Pseudoxanthoma Elasticum/complications , Pseudoxanthoma Elasticum/enzymology , Pyrophosphatases/genetics , Vascular Calcification/complications , Zebrafish/genetics , Animals , Biomarkers/metabolism , Calcinosis/drug therapy , Calcinosis/enzymology , Calcium/metabolism , Choristoma/enzymology , Choristoma/pathology , Etidronic Acid/pharmacology , Etidronic Acid/therapeutic use , Fibroblast Growth Factor-23 , Homeostasis/drug effects , Humans , Notochord/drug effects , Notochord/pathology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Phenotype , Phosphates/metabolism , Pseudoxanthoma Elasticum/drug therapy , Vascular Calcification/drug therapy , Vascular Calcification/enzymology
14.
Development ; 141(11): 2225-34, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24821985

ABSTRACT

Zebrafish have an unlimited capacity to regenerate bone after fin amputation. In this process, mature osteoblasts dedifferentiate to osteogenic precursor cells and thus represent an important source of newly forming bone. By contrast, differentiated osteoblasts do not appear to contribute to repair of bone injuries in mammals; rather, osteoblasts form anew from mesenchymal stem cells. This raises the question whether osteoblast dedifferentiation is specific to appendage regeneration, a special feature of the lepidotrichia bone of the fish fin, or a process found more generally in fish bone. Here, we show that dedifferentiation of mature osteoblasts is not restricted to fin regeneration after amputation, but also occurs during repair of zebrafish fin fractures and skull injuries. In both models, mature osteoblasts surrounding the injury downregulate the expression of differentiation markers, upregulate markers of the pre-osteoblast state and become proliferative. Making use of photoconvertible Kaede protein as well as Cre-driven genetic fate mapping, we show that osteoblasts migrate to the site of injury to replace damaged tissue. Our findings suggest a fundamental role for osteoblast dedifferentiation in reparative bone formation in fish and indicate that adult fish osteoblasts display elevated cellular plasticity compared with mammalian bone-forming cells.


Subject(s)
Animal Fins/pathology , Bone and Bones/injuries , Bone and Bones/pathology , Cell Differentiation , Osteoblasts/cytology , Skull/pathology , Animals , Animals, Genetically Modified , Bone Regeneration , Cell Proliferation , Fluorescent Dyes , Gene Expression Regulation , Green Fluorescent Proteins/chemistry , Luminescent Proteins/metabolism , Necrosis , Osteoblasts/metabolism , Osteogenesis/drug effects , Wound Healing , Zebrafish
15.
PLoS One ; 9(3): e90568, 2014.
Article in English | MEDLINE | ID: mdl-24608635

ABSTRACT

The extracellular matrix of the immature and mature skeleton is key to the development and function of the skeletal system. Notwithstanding its importance, it has been technically challenging to obtain a comprehensive picture of the changes in skeletal composition throughout the development of bone and cartilage. In this study, we analyzed the extracellular protein composition of the zebrafish skeleton using a mass spectrometry-based approach, resulting in the identification of 262 extracellular proteins, including most of the bone and cartilage specific proteins previously reported in mammalian species. By comparing these extracellular proteins at larval, juvenile, and adult developmental stages, 123 proteins were found that differed significantly in abundance during development. Proteins with a reported function in bone formation increased in abundance during zebrafish development, while analysis of the cartilage matrix revealed major compositional changes during development. The protein list includes ligands and inhibitors of various signaling pathways implicated in skeletogenesis such as the Int/Wingless as well as the insulin-like growth factor signaling pathways. This first proteomic analysis of zebrafish skeletal development reveals that the zebrafish skeleton is comparable with the skeleton of other vertebrate species including mammals. In addition, our study reveals 6 novel proteins that have never been related to vertebrate skeletogenesis and shows a surprisingly large number of differences in the cartilage and bone proteome between the head, axis and caudal fin regions. Our study provides the first systematic assessment of bone and cartilage protein composition in an entire vertebrate at different stages of development.


Subject(s)
Extracellular Matrix/metabolism , Proteomics/methods , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals
16.
Proc Natl Acad Sci U S A ; 109(52): 21372-7, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23236130

ABSTRACT

Bone mineralization is an essential step during the embryonic development of vertebrates, and bone serves vital functions in human physiology. To systematically identify unique gene functions essential for osteogenesis, we performed a forward genetic screen in zebrafish and isolated a mutant, no bone (nob), that does not form any mineralized bone. Positional cloning of nob identified the causative gene to encode ectonucleoside triphosphate/diphosphohydrolase 5 (entpd5); analysis of its expression pattern demonstrates that entpd5 is specifically expressed in osteoblasts. An additional mutant, dragonfish (dgf), exhibits ectopic mineralization in the craniofacial and axial skeleton and encodes a loss-of-function allele of ectonucleotide pyrophosphatase phosphodiesterase 1 (enpp1). Intriguingly, generation of double-mutant nob/dgf embryos restored skeletal mineralization in nob mutants, indicating that mechanistically, Entpd5 and Enpp1 act as reciprocal regulators of phosphate/pyrophosphate homeostasis in vivo. Consistent with this, entpd5 mutant embryos can be rescued by high levels of inorganic phosphate, and phosphate-regulating factors, such as fgf23 and npt2a, are significantly affected in entpd5 mutant embryos. Our study demonstrates that Entpd5 represents a previously unappreciated essential player in phosphate homeostasis and skeletal mineralization.


Subject(s)
Calcification, Physiologic , Homeostasis , Phosphates/metabolism , Pyrophosphatases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Alleles , Amino Acid Sequence , Animals , Base Sequence , Bone and Bones/embryology , Bone and Bones/metabolism , Bone and Bones/pathology , Embryo, Nonmammalian/metabolism , Fibroblast Growth Factor-23 , Humans , Molecular Sequence Data , Mutation/genetics , Organ Specificity , Osteoblasts/enzymology , Phenotype , Phosphoric Diester Hydrolases/genetics , Pyrophosphatases/chemistry , Pyrophosphatases/genetics , Zebrafish/embryology , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
17.
FASEB J ; 26(10): 4092-101, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22787265

ABSTRACT

To systematically identify novel gene functions essential for osteogenesis and skeletal mineralization, we performed a forward genetic mutagenesis screen in zebrafish and isolated a mutant that showed delayed skeletal mineralization. Analysis of the mutant phenotype in an osterix:nuclear-GFP transgenic background demonstrated that mutants contain osterix-expressing osteoblasts comparable to wild-type embryos. Positional cloning revealed a premature stop mutation in the macrophage-stimulating protein (msp) gene, predicted to result in a biologically inactive protein. Analysis of the embryonic expression pattern for the receptor for Msp, Ron, shows specific expression in the corpuscles of Stannius, a teleost-specific organ that produces stanniocalcin, a pivotal hormone in fish calcium homeostasis. Knockdown of Ron resulted in identical phenotypes as observed in msp mutants. Msp mutant embryos could be rescued by excess calcium. Consistent with a role for Msp/Ron in calcium homeostasis, calcium-regulating factors, such as pth1, pth2, stc1l, and trpv5/6 were significantly affected in msp mutant larvae. While Msp and Ron have previously been shown to play a critical role in a wide variety of biological processes, we introduce here the Msp/Ron signaling axis as a previously unappreciated player in calcium homeostasis and embryonic skeletal mineralization.


Subject(s)
Calcium/metabolism , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Glycoproteins/metabolism , Hepatocyte Growth Factor/genetics , Homeostasis/genetics , Homeostasis/physiology , Osteogenesis/genetics , Osteogenesis/physiology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics
18.
FASEB J ; 25(9): 3197-207, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21670068

ABSTRACT

Calcium is an essential ion serving a multitude of physiological roles. Aside from its role as a second messenger, it is an essential component of the vertebrate bone matrix. Efficient uptake and storage of calcium are therefore indispensable for all vertebrates. Transient receptor potential family, vanilloid type (TRPV)5 and TRPV6 channels are known players in transcellular calcium uptake, but the exact contribution of this pathway is unclear. We used forward genetic screening in zebrafish (Danio rerio) to identify genes essential in bone formation and identified a lethal zebrafish mutant (matt-und-schlapp) with severe defects in bone formation, including lack of ossification of the vertebral column and craniofacial structures. Mutant embryos show a 68% reduction in calcium content, and systemic calcium homeostasis is disturbed when compared with siblings. The phenotype can be partially rescued by increasing ambient calcium levels to 25 mM. We identified the mutation as a loss-of-function mutation in the single orthologue of TRPV5 and 6, trpv5/6. Expression in HEK293 cells showed that Trpv5/6 is a calcium-selective channel capable of inward calcium transport at physiological concentrations whereas the mutant channel is not. Taken together, this study provides both genetic and functional evidence that transcellular epithelial calcium uptake is vital to sustain life and enable bone formation.


Subject(s)
Bone Development/physiology , Calcium/metabolism , Epithelium/embryology , Epithelium/metabolism , TRPV Cation Channels/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Bone Development/genetics , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/physiology , HEK293 Cells , Humans , Molecular Sequence Data , Mutation , TRPV Cation Channels/genetics , Zebrafish Proteins/genetics
19.
Biochim Biophys Acta ; 1774(9): 1108-17, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17669701

ABSTRACT

During biomineralization the organism controls the nature, orientation, size and shape of the mineral phase. The aim of this study was to investigate whether proteins or vesicles that are constitutively released by growing ATDC5 cells have the ability to affect the formation of the calcium phosphate crystal. Therefore, subconfluent cultured ATDC5 cells were incubated for 1 h in medium without serum. Subsequently, medium was harvested and incubated for 24 h in the presence of additional Pi. This resulted in the formation of flat mineralizing structures (FMS), consisting of complex irregularly shaped flat crystals, which occasionally contained fiber-like structures ( approximately 40 microm in size). Without pre-incubation of medium with cells, only small punctate (dot like) calcium phosphate precipitates were observed. The formation of FMS was shown to be caused by soluble factors released by subconfluent ATDC5 cells. Proteomic analysis by mass spectrometry showed that FMS contained a specific set intracellular proteins, serum proteins, and extracellular matrix proteins. Bulk cytosolic proteins derived from homogenized cells or serum proteins did, however, not induce the formation of FMS. Conditioned medium from HeLa, CHO K1, RAW 264.7 and MDCK cells was also capable to form FMS under our experimental conditions. Therefore the formation of FMS seems to be caused by specific soluble factors constitutively released by ADTC5 and other cells. This in vitro model system can be used as a tool to identify factors that affect the shape of the biomineral phase.


Subject(s)
Calcium Phosphates/chemistry , Chondrocytes/metabolism , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Crystallization , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Mice , Microscopy, Electron , Proteins/physiology , Spectrophotometry, Infrared
20.
Front Biosci ; 12: 2631-45, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17127268

ABSTRACT

Mineralization is an essential requirement for normal skeletal development, but under certain pathological conditions organs like articular cartilage and cardiovascular tissue are prone to unwanted mineralization. Recent findings suggest that the mechanisms regulating skeletal mineralization may be similar to those regulating pathological mineralization. In general, three forms of cell-mediated mineralization are recognized in an organism: intramembranous ossification, endochondral ossification and pathological mineralization. This review summarizes recent work that tried to elucidate how cell-mediated mineralization is initiated and regulated. To explain mineralization, several theories have been proposed. One theory proposes that mineralization is initiated within matrix vesicles (MVs). A second, not mutually exclusive, theory proposes that phosphate induces apoptosis, and that apoptotic bodies nucleate crystals composed of calcium and phosphate. A third theory suggests that mineralization is mediated by certain non-collagenous proteins, which associate with the extracellular matrix. Regardless of the way mineralization is initiated, the organism also actively inhibits mineralization by specific proteins and removal of an inhibitor may also induce mineralization. Although many studies greatly contributed to a better understanding of the mechanisms regulating cell-mediated mineralization, many questions remain about the mechanisms that trigger cell-mediated mineralization and how this process is regulated. Further investigation is necessary to develop in the future novel therapeutic strategies to prevent pathological mineralization.


Subject(s)
Calcification, Physiologic , Animals , Apoptosis , Calcinosis/etiology , Cytoplasmic Vesicles/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/physiology , Humans , Mice , alpha-Fetoproteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...