Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Dis Model Mech ; 14(3)2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33563616

RESUMO

Collagen assembly during development is essential for successful matrix mineralisation, which determines bone quality and mechanocompetence. However, the biochemical and structural perturbations that drive pathological skeletal collagen configuration remain unclear. Deletion of vascular endothelial growth factor (VEGF; also known as VEGFA) in bone-forming osteoblasts (OBs) induces sex-specific alterations in extracellular matrix (ECM) conformation and mineralisation coupled to vascular changes, which are augmented in males. Whether this phenotypic dimorphism arises as a result of the divergent control of ECM composition and its subsequent arrangement is unknown and is the focus of this study. Herein, we used murine osteocalcin-specific Vegf knockout (OcnVEGFKO) and performed ex vivo multiscale analysis at the tibiofibular junction of both sexes. Label-free and non-destructive polarisation-resolved second-harmonic generation (p-SHG) microscopy revealed a reduction in collagen fibre number in males following the loss of VEGF, complemented by observable defects in matrix organisation by backscattered electron scanning electron microscopy. This was accompanied by localised divergence in collagen orientation, determined by p-SHG anisotropy measurements, as a result of OcnVEGFKO. Raman spectroscopy confirmed that the effect on collagen was linked to molecular dimorphic VEGF effects on collagen-specific proline and hydroxyproline, and collagen intra-strand stability, in addition to matrix carbonation and mineralisation. Vegf deletion in male and female murine OB cultures in vitro further highlighted divergence in genes regulating local ECM structure, including Adamts2, Spp1, Mmp9 and Lama1. Our results demonstrate the utility of macromolecular imaging and spectroscopic modalities for the detection of collagen arrangement and ECM composition in pathological bone. Linking the sex-specific genetic regulators to matrix signatures could be important for treatment of dimorphic bone disorders that clinically manifest in pathological nano- and macro-level disorganisation. This article has an associated First Person interview with the first author of the paper.


Assuntos
Matriz Extracelular , Fator A de Crescimento do Endotélio Vascular , Animais , Osso e Ossos/metabolismo , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Feminino , Humanos , Masculino , Camundongos , Osteoblastos , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Ann Integr Mol Med ; 1(1): 23-47, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31656957

RESUMO

Our initial unidirectional understanding of the flow of protein-encoding genetic information, DNA to RNA to protein, a process defined as the "Central Dogma of Molecular Biology" and usually depicted as a downward arrow, was eventually amended to account for the "vertical" information back-flow from RNA to DNA, reverse transcription, and for its "horizontal" side-flow from RNA to RNA, RNA-dependent RNA synthesis, RdRs. These processes, both potentially leading to protein production, were assumed to be strictly virus-specific. However, whereas this presumption might be true for the former, it became apparent that the cellular enzymatic machinery for the later, a conventional RNA-dependent RNA polymerase activity, RdRp, is ubiquitously present and RdRs regularly occurs in eukaryotes. The strongest evidence for the occurrence and functionality of RdRp activity in mammalian cells comes from viruses, such as hepatitis delta virus, HDV, that do not encode RdRp yet undergo a robust RNA replication once inside the host. Eventually, it became clear that RdRp activity, apparently in a non-conventional form, is constitutively present in most, if not in all, mammalian cells. Because such activity was shown to produce short transcripts, because of its apparent involvement in RNA interference phenomena, and because double-stranded RNA is known to trigger cellular responses leading to its degradation, it was generally assumed that its role in mammalian cells is restricted to a regulatory function. However, at the same time, an enzymatic activity capable of generating complete antisense RNA complements of mRNAs was discovered in mammalian cells undergoing terminal differentiation. Moreover, observations of widespread synthesis of antisense RNA initiating at the 3'poly(A) of mRNAs in human cells suggested an extensive cellular utilization of mammalian RdRp. These results led to the development of a model of RdRp-facilitated and antisense RNA-mediated amplification of mammalian mRNA. Here, we report the in vivo detection in cells undergoing terminal erythroid differentiation of the major model-predicted identifiers of such a process, a chimeric double-stranded/pinhead-structured intermediates containing both sense and antisense RNA strands covalently joined in a rigorously predicted and uniquely defined manner. We also report the identification of the putative chimeric RNA end product of mRNA amplification. It is heavily modified, uniformly truncated, yet retains the intact coding region, and terminates with the OH group at both ends; its massive cellular amount is unprecedented for a conventional mRNA transcription product and it translates into polypeptides indistinguishable from the translation product of conventional mRNA. Moreover, we describe the occurrence of the second Tier of mammalian RNA-dependent mRNA amplification, a physiologically occurring, RdRp-driven intracellular PCR process, "iPCR", and report the detection of its distinct RNA end products. Whether mammalian mRNA amplification is a specialized occurrence limited to extreme circumstances of terminal differentiation in cells programmed for only a short survival span or a general physiological phenomenon was answered in the companion article Volloch et al. Ann Integr Mol Med. 2019;1(1):1004. by the detection of major identifiers of this process for mRNA encoding α1, ß1, and γ1 chains of laminin, a major extracellular matrix protein abundantly produced throughout the tissue and organ development and homeostasis and an exceptionally revealing indicator of the range and scope of this phenomenon. The results obtained introduce the occurrence of RNA-dependent mRNA amplification as a new mode of genomic protein-encoding information transfer in mammalian cells and establish it as a general physiological phenomenon.

3.
J Bone Miner Res ; 34(11): 2117-2132, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31269275

RESUMO

Osteoblast (OB) lineage cells are an important source of vascular endothelial growth factor (VEGF), which is critical for bone growth and repair. During bone development, pubertal differences in males and females exist, but little is known about whether VEGF signaling contributes to skeletal sexual dimorphism. We have found that in mice, conditional disruption of VEGF in osteocalcin-expressing cells (OcnVEGFKO) exerts a divergent influence on morphological, cellular, and whole bone properties between sexes. Furthermore, we describe an underlying sexual divergence in VEGF signaling in OB cultures in vitro independent of circulating sex hormones. High-resolution synchrotron computed tomography and backscattered scanning electron microscopy revealed, in males, extensive unmineralized osteoid encasing enlarged blood vessel canals and osteocyte lacunae in cortical bone after VEGF deletion, which contributed to increased porosity. VEGF was deleted in male and female long bone-derived OBs (OBVEGKO) in vitro and Raman spectroscopic analyses of mineral and matrix repertoires highlighted differences between male and female OBVEGFKO cells, with increased immature phosphate species prevalent in male OBVEGFKO cultures versus wild type (WT). Further sexual dimorphism was observed in bone marrow endothelial cell gene expression in vitro after VEGF deletion and in sclerostin protein expression, which was increased in male OcnVEGFKO bones versus WT. The impact of altered OB matrix composition after VEGF deletion on whole bone geometry was assessed between sexes, although significant differences between OcnVEGFKO and WT were identified only in females. Our results suggest that bone-derived VEGF regulates matrix mineralization and vascularization distinctly in males and females, which results in divergent physical bone traits.


Assuntos
Desenvolvimento Ósseo , Células da Medula Óssea/metabolismo , Osso e Ossos/irrigação sanguínea , Células Endoteliais/metabolismo , Caracteres Sexuais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Osso e Ossos/metabolismo , Feminino , Masculino , Camundongos , Camundongos Knockout , Fator A de Crescimento do Endotélio Vascular/genética
4.
Sci Rep ; 7(1): 13027, 2017 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-29026147

RESUMO

Genome wide studies indicate that vascular endothelial growth factor A (VEGF) is associated with osteoarthritis (OA), and increased VEGF expression correlates with increased disease severity. VEGF is also a chondrocyte survival factor during development and essential for bone formation, skeletal growth and postnatal homeostasis. This raises questions of how the important embryonic and postnatal functions of VEGF can be reconciled with an apparently destructive role in OA. Addressing these questions, we find that VEGF acts as a survival factor in growth plate chondrocytes during development but only up until a few weeks after birth in mice. It is also required for postnatal differentiation of articular chondrocytes and the timely ossification of bones in joint regions. In surgically induced knee OA in mice, a model of post-traumatic OA in humans, increased expression of VEGF is associated with catabolic processes in chondrocytes and synovial cells. Conditional knock-down of Vegf attenuates induced OA. Intra-articular anti-VEGF antibodies suppress OA progression, reduce levels of phosphorylated VEGFR2 in articular chondrocytes and synovial cells and reduce levels of phosphorylated VEGFR1 in dorsal root ganglia. Finally, oral administration of the VEGFR2 kinase inhibitor Vandetanib attenuates OA progression.


Assuntos
Cartilagem Articular/embriologia , Cartilagem Articular/patologia , Osteoartrite/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Administração Oral , Animais , Anticorpos/farmacologia , Desenvolvimento Ósseo , Diferenciação Celular , Linhagem da Célula , Condrócitos/metabolismo , Colágeno Tipo II/metabolismo , Progressão da Doença , Endotélio/metabolismo , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Lâmina de Crescimento/metabolismo , Lâmina de Crescimento/patologia , Integrases/metabolismo , Articulação do Joelho/patologia , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Osteoartrite/patologia , Osteogênese , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Fator A de Crescimento do Endotélio Vascular/deficiência , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
5.
Matrix Biol ; 62: 105-114, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28011198

RESUMO

Our previous studies of Antxr1 knockout mice suggested that fibrotic skin abnormalities in these mice are associated with increased VEGF signaling. Here, based on studies of primary fibroblasts isolated from skin of Antx1+/+ and Antxr1-/- mice at embryonic stage E17.5 and postnatal day P49, we conclude that increased Col1a1 and Fn1 expression in Antxr1-deficient fibroblasts is partly mediated by a cell-autonomous ANTXR1-dependent mechanism. In turn, this may act in parallel with VEGF-dependent regulation of collagen type I and fibronectin production. We demonstrate that shRNA mediated knockdown of VEGF in Antxr1-/- fibroblasts reduces Col1a1 and Fn1 expression to below control levels, and these are restored by exogenous addition of recombinant VEGF. In addition, the increase in protein levels of collagen type I and fibronectin in mutant cells is blocked by VEGF neutralizing antibody. However, expressing the longest isoform of ANTXR1 (sv1) in mutant fibroblasts decreases levels of Ctgf, Col1a1 and Fn1 transcripts, but has no effect on VEGF expression. Taken together, our data suggest that the increased matrix production in Antxr1- deficient fibroblasts primarily occurs via a CTGF-dependent pathway and that other ANTXR1-associated mechanisms contribute to VEGF-dependent increase of collagen type I and fibronectin expression. Our findings provide a basis for further studies of novel ANTXR1-dependent connective tissue homeostatic control mechanisms in healthy individuals, patients with organ fibrosis, and patients with GAPO syndrome.


Assuntos
Biomarcadores Tumorais/genética , Colágeno Tipo I/metabolismo , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Receptores de Peptídeos/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Células Cultivadas , Cadeia alfa 1 do Colágeno Tipo I , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Matriz Extracelular/metabolismo , Técnicas de Inativação de Genes , Homeostase , Camundongos , Proteínas dos Microfilamentos , Receptores de Superfície Celular , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
J Orthop Res ; 35(7): 1461-1469, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27658810

RESUMO

BMP2 is widely used for promotion of bone repair and regeneration. However, bone formation induced by BMP2 is quite variable. Bone forming progenitor cells in different locations appear to respond to BMP2 in different ways, and repair outcomes can vary as a consequence of modulating effects by other factors. In this study, we have examined the effects of VEGF on BMP2-induced repair of a cortical bone defect, a 1 mm diameter drill hole, in the proximal tibia of mice. Treatment of the defect with either a bolus of PBS or soluble VEGFR1 (sVEGFR1), a decoy receptor for VEGF, had the same effects on bone formation via intramembranous ossification in the defect and cartilage formation and injured periosteum, during the healing process. In contrast, treatment with BMP2 inhibited intramembranous bone formation in the defect while it promoted cartilage and endochondral bone formation in the injured periosteum compared with mice treated with PBS or sVEGFR1. The inhibitory effect of BMP2 on bone formation was unlikely due to increased osteoclast activity and decreased invasion of blood vessels in the defect. Most importantly, co-delivery of BMP2 and sVEGFR1 reversed the inhibition of intramembranous bone formation by BMP2. Furthermore, the decreased accumulation of collagen and production of bone matrix proteins in the defect of groups with BMP2 treatment could also be prevented by co-delivery of BMP2 and sVEGFR1. Our data indicate that introducing a VEGF-binding protein, such as sVEGFR1, to reduce levels of extracellular VEGF, may enhance the effects of BMP2 on intramembranous bone formation. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1461-1469, 2017.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Consolidação da Fratura/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteína Morfogenética Óssea 2/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Fraturas Ósseas/tratamento farmacológico , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Transgênicos , Neovascularização Fisiológica/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/uso terapêutico
7.
Dev Dyn ; 246(4): 227-234, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27750398

RESUMO

Vascular endothelial growth factor A (VEGF) is a critical regulator of vascular development and postnatal angiogenesis and homeostasis, and it is essential for bone development and repair. Blood vessels serve both as structural templates for bone formation and they provide essential cells, growth factors and minerals needed for synthesis and mineralization, as well as turnover, of the extracellular matrix in bone. Through its regulation of angiogenesis, VEGF contributes to coupling of osteogenesis to angiogenesis, and it directly controls the differentiation and function of osteoblasts and osteoclasts. In this review, we summarize the properties of VEGF and its receptors that are relevant to bone formation and repair; the roles of VEGF during development of endochondral and membranous bones; and the contributions of VEGF to bone healing during different phases of bone repair. Finally, we discuss contributions of altered VEGF function in inherited disorders with bone defects as part of their phenotypes, and we speculate on what will be required before therapeutic strategies based on VEGF modulation can be developed for clinical use to treat patients with bone growth disorders and/or compromised bone repair. Developmental Dynamics 246:227-234, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Desenvolvimento Ósseo , Regeneração Óssea , Fator A de Crescimento do Endotélio Vascular/fisiologia , Fatores de Crescimento do Endotélio Vascular/fisiologia , Animais , Doenças do Desenvolvimento Ósseo/congênito , Remodelação Óssea , Osso e Ossos , Humanos
8.
Int J Exp Pathol ; 97(4): 296-302, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27581728

RESUMO

Studies of proliferative hemangiomas have led to the discovery that interactions of endothelial cells with extracellular matrix and/or Vascular Endothelial Growth Factor (VEGF)-A stimulate the expression of VEGFR1, the VEGF decoy receptor, and suppress VEGF-dependent VEGFR2 signalling by a mechanism that requires the matrix-binding receptor Anthrax Toxin Receptor (ANTXR)1, VEGFR2, ß1 integrin and the Nuclear Factor of Activated T cells (NFAT). In hemangioma endothelial cells, all these components are present, but are functionally compromised, so that the levels of VEGFR1 are extremely low and VEGFR2 signalling is constitutively active. Consequently, the levels of Hypoxia Inducible Factor (HIF)-1α and its transcriptional targets, VEGF-A and C-X-C motif chemokine 12 (CxCl12), are elevated and a positive VEGF-A feedback loop is established. Overexpression of ANTXR1, carrying a heterozygous Ala-to-Thr mutation, induces hemangioma-like signalling in control endothelial cells; VEGF signalling is normalized when wild-type ANTXR1 is overexpressed in hemangioma cells. These findings suggest that ANTXR1 functions as a negative regulator of VEGF-A signalling. Studies of a mouse model of the Growth Retardation, Alopecia, Pseudo-anodontia and Optic Atrophy (GAPO) syndrome, caused by the loss-of-function mutations in ANTXR1, as well as knock-in mice carrying the Ala-to-Thr ANTXR1 mutation, confirm that ANTXR1 functions as a suppressor of VEGF-A signalling. Cutaneous endothelial cells isolated from ANTXR1-deficient mice exhibit low levels of VEGFR1, elevated levels of VEGF-A, HIF-1α and CxCl12 and activated VEGFR2 signalling as in hemangioma. Increased numbers of myeloid cells in the skin of ANTXR1-deficient mice are associated with reduced vascularity and increased skin fibrosis, suggesting a mechanism for hemangioma involution and replacement by fibrotic scars. Through controlling VEGF-A signalling and extracellular matrix synthesis, ANTXR1 is emerging as a key regulator of skeletal and connective tissue development and homeostasis.


Assuntos
Desenvolvimento Ósseo/fisiologia , Tecido Conjuntivo/crescimento & desenvolvimento , Hemangioma/metabolismo , Homeostase/fisiologia , Animais , Hemangioma/patologia , Humanos , Proteínas dos Microfilamentos , Proteínas de Neoplasias/fisiologia , Receptores de Superfície Celular/fisiologia , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
9.
Bone ; 91: 30-8, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27353702

RESUMO

Vascular endothelial growth factor-A (VEGF) is one of the most important growth factors for regulation of vascular development and angiogenesis. Since bone is a highly vascularized organ and angiogenesis plays an important role in osteogenesis, VEGF also influences skeletal development and postnatal bone repair. Compromised bone repair and regeneration in many patients can be attributed to impaired blood supply; thus, modulation of VEGF levels in bones represents a potential strategy for treating compromised bone repair and improving bone regeneration. This review (i) summarizes the roles of VEGF at different stages of bone repair, including the phases of inflammation, endochondral ossification, intramembranous ossification during callus formation and bone remodeling; (ii) discusses different mechanisms underlying the effects of VEGF on osteoblast function, including paracrine, autocrine and intracrine signaling during bone repair; (iii) summarizes the role of VEGF in the bone regenerative procedure, distraction osteogenesis; and (iv) reviews evidence for the effects of VEGF in the context of repair and regeneration techniques involving the use of scaffolds, skeletal stem cells and growth factors.


Assuntos
Regeneração Óssea , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização , Animais , Humanos , Osteoblastos/metabolismo , Osteogênese , Osteogênese por Distração
10.
J Bone Miner Res ; 31(5): 911-24, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27163679

RESUMO

Increased vascular endothelial growth factor (VEGF) levels are associated with osteoarthritis (OA) progression. Indeed, VEGF appears to be involved in OA-specific pathologies including cartilage degeneration, osteophyte formation, subchondral bone cysts and sclerosis, synovitis, and pain. Moreover, a wide range of studies suggest that inhibition of VEGF signaling reduces OA progression. This review highlights both the potential significance of VEGF in OA pathology and pain, as well as potential benefits of inhibition of VEGF and its receptors as an OA treatment. With the emergence of the clinical use of anti-VEGF therapy outside of OA, both as high-dose systemic treatments and low-dose local treatments, these particular therapies are now more widely understood. Currently, there is no established disease-modifying drug available for patients with OA, which warrants continued study of the inhibition of VEGF signaling in OA, as stand-alone or adjuvant therapy. © 2016 American Society for Bone and Mineral Research.


Assuntos
Osteoartrite/tratamento farmacológico , Dor/tratamento farmacológico , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Feminino , Humanos , Masculino , Osteoartrite/metabolismo , Dor/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Matrix Biol ; 52-54: 127-140, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26899202

RESUMO

Deficiency of vascular endothelial growth factor A (VEGF) has been associated with severe craniofacial anomalies in both humans and mice. Cranial neural crest cell (NCC)-derived VEGF regulates proliferation, vascularization and ossification of cartilage and membranous bone. However, the function of VEGF derived from specific subpopulations of NCCs in controlling unique aspects of craniofacial morphogenesis is not clear. In this study a conditional knockdown strategy was used to genetically delete Vegfa expression in Osterix (Osx) and collagen II (Col2)-expressing NCC descendants. No major defects in calvaria and mandibular morphogenesis were observed upon knockdown of VEGF in the Col2(+) cell population. In contrast, loss of VEGF in Osx(+) osteoblast progenitor cells led to reduced ossification of calvarial and mandibular bones without affecting the formation of cartilage templates in newborn mice. The early stages of ossification in the developing jaw revealed decreased initial mineralization levels and a reduced thickness of the collagen I (Col1)-positive bone template upon loss of VEGF in Osx(+) precursors. Increased numbers of proliferating cells were detected within the jaw mesenchyme of mutant embryos. Explant culture assays revealed that mandibular osteogenesis occurred independently of paracrine VEGF action and vascular development. Reduced VEGF expression in mandibles coincided with increased phospho-Smad1/5 (P-Smad1/5) levels and bone morphogenetic protein 2 (Bmp2) expression in the jaw mesenchyme. We conclude that VEGF derived from Osx(+) osteoblast progenitor cells is required for optimal ossification of developing mandibular bones and modulates mechanisms controlling BMP-dependent specification and expansion of the jaw mesenchyme.


Assuntos
Mandíbula/crescimento & desenvolvimento , Crista Neural/citologia , Crânio/crescimento & desenvolvimento , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Recém-Nascidos , Calcificação Fisiológica , Proliferação de Células , Células Cultivadas , Colágeno Tipo II/metabolismo , Técnicas de Silenciamento de Genes , Mandíbula/metabolismo , Camundongos , Crânio/metabolismo , Fator de Transcrição Sp7/metabolismo
12.
J Clin Invest ; 126(2): 509-26, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26731472

RESUMO

Osteoblast-derived VEGF is important for bone development and postnatal bone homeostasis. Previous studies have demonstrated that VEGF affects bone repair and regeneration; however, the cellular mechanisms by which it works are not fully understood. In this study, we investigated the functions of osteoblast-derived VEGF in healing of a bone defect. The results indicate that osteoblast-derived VEGF plays critical roles at several stages in the repair process. Using transgenic mice with osteoblast-specific deletion of Vegfa, we demonstrated that VEGF promoted macrophage recruitment and angiogenic responses in the inflammation phase, and optimal levels of VEGF were required for coupling of angiogenesis and osteogenesis in areas where repair occurs by intramembranous ossification. VEGF likely functions as a paracrine factor in this process because deletion of Vegfr2 in osteoblastic lineage cells enhanced osteoblastic maturation and mineralization. Furthermore, osteoblast- and hypertrophic chondrocyte-derived VEGF stimulated recruitment of blood vessels and osteoclasts and promoted cartilage resorption at the repair site during the periosteal endochondral ossification stage. Finally, osteoblast-derived VEGF stimulated osteoclast formation in the final remodeling phase of the repair process. These findings provide a basis for clinical strategies to improve bone regeneration and treat defects in bone healing.


Assuntos
Regeneração Óssea/fisiologia , Calcificação Fisiológica/fisiologia , Diferenciação Celular/fisiologia , Osteoblastos/metabolismo , Osteogênese/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Camundongos , Camundongos Knockout , Osteoblastos/citologia , Fator A de Crescimento do Endotélio Vascular/genética
13.
Proc Natl Acad Sci U S A ; 113(3): E338-47, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26721400

RESUMO

Pathologic extraskeletal bone formation, or heterotopic ossification (HO), occurs following mechanical trauma, burns, orthopedic operations, and in patients with hyperactivating mutations of the type I bone morphogenetic protein receptor ACVR1 (Activin type 1 receptor). Extraskeletal bone forms through an endochondral process with a cartilage intermediary prompting the hypothesis that hypoxic signaling present during cartilage formation drives HO development and that HO precursor cells derive from a mesenchymal lineage as defined by Paired related homeobox 1 (Prx). Here we demonstrate that Hypoxia inducible factor-1α (Hif1α), a key mediator of cellular adaptation to hypoxia, is highly expressed and active in three separate mouse models: trauma-induced, genetic, and a hybrid model of genetic and trauma-induced HO. In each of these models, Hif1α expression coincides with the expression of master transcription factor of cartilage, Sox9 [(sex determining region Y)-box 9]. Pharmacologic inhibition of Hif1α using PX-478 or rapamycin significantly decreased or inhibited extraskeletal bone formation. Importantly, de novo soft-tissue HO was eliminated or significantly diminished in treated mice. Lineage-tracing mice demonstrate that cells forming HO belong to the Prx lineage. Burn/tenotomy performed in lineage-specific Hif1α knockout mice (Prx-Cre/Hif1α(fl:fl)) resulted in substantially decreased HO, and again lack of de novo soft-tissue HO. Genetic loss of Hif1α in mesenchymal cells marked by Prx-cre prevents the formation of the mesenchymal condensations as shown by routine histology and immunostaining for Sox9 and PDGFRα. Pharmacologic inhibition of Hif1α had a similar effect on mesenchymal condensation development. Our findings indicate that Hif1α represents a promising target to prevent and treat pathologic extraskeletal bone.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Ossificação Heterotópica/genética , Ossificação Heterotópica/prevenção & controle , Ferimentos e Lesões/complicações , Receptores de Ativinas Tipo I/metabolismo , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Queimaduras/complicações , Queimaduras/genética , Condrogênese/efeitos dos fármacos , Condrogênese/genética , Modelos Animais de Doenças , Redes Reguladoras de Genes/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Integrases/metabolismo , Medições Luminescentes , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos Knockout , Modelos Biológicos , Compostos de Mostarda/farmacologia , Ossificação Heterotópica/diagnóstico por imagem , Ossificação Heterotópica/tratamento farmacológico , Fenilpropionatos/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fatores de Transcrição SOX9/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Tendões/efeitos dos fármacos , Tendões/patologia , Tendões/cirurgia , Tenotomia , Regulação para Cima/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Ferimentos e Lesões/patologia , Microtomografia por Raio-X
14.
J Biomed Opt ; 20(8): 80501, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26263412

RESUMO

Calciphylaxis is a painful, debilitating, and premorbid condition, which presents as calcified vasculature and soft tissues. Traditional diagnosis of calciphylaxis lesions requires an invasive biopsy, which is destructive, time consuming, and often leads to exacerbation of the condition and infection. Furthermore, it is difficult to find small calcifications within a large wound bed. To address this need, a noninvasive diagnostic tool may help clinicians identify ectopic calcified mineral and determine the disease margin. We propose Raman spectroscopy as a rapid, point-of-care, noninvasive, and label-free technology to detect calciphylaxis mineral. Debrided calciphylactic tissue was collected from six patients and assessed by microcomputed tomography (micro-CT). Micro-CT confirmed extensive deposits in three specimens, which were subsequently examined with Raman spectroscopy. Raman spectra confirmed that deposits were consistent with carbonated apatite, consistent with the literature. Raman spectroscopy shows potential as a noninvasive technique to detect calciphylaxis in a clinical environment.


Assuntos
Apatitas/metabolismo , Calciofilaxia/diagnóstico , Calciofilaxia/metabolismo , Cálcio/metabolismo , Sensibilidade e Especificidade , Análise Espectral Raman/métodos , Biomarcadores/metabolismo , Humanos , Reprodutibilidade dos Testes , Coloração e Rotulagem
15.
Development ; 142(11): 1984-91, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25977369

RESUMO

Vascular endothelial growth factor A (Vegfa) has important roles in endochondral bone formation. Osteoblast precursors, endothelial cells and osteoclasts migrate from perichondrium into primary ossification centers of cartilage templates of future bones in response to Vegfa secreted by (pre)hypertrophic chondrocytes. Perichondrial osteolineage cells also produce Vegfa, but its function is not well understood. By deleting Vegfa in osteolineage cells in vivo, we demonstrate that progenitor-derived Vegfa is required for blood vessel recruitment in perichondrium and the differentiation of osteoblast precursors in mice. Conditional deletion of Vegfa receptors indicates that Vegfa-dependent effects on osteoblast differentiation are mediated by Vegf receptor 2 (Vegfr2). In addition, Vegfa/Vegfr2 signaling stimulates the expression and activity of Indian hedgehog, increases the expression of ß-catenin and inhibits Notch2. Our findings identify Vegfa as a regulator of perichondrial vascularity and osteoblast differentiation at early stages of bone development.


Assuntos
Desenvolvimento Ósseo , Osso e Ossos/irrigação sanguínea , Diferenciação Celular , Neovascularização Fisiológica , Osteoblastos/citologia , Osteoblastos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Osso e Ossos/metabolismo , Calcificação Fisiológica , Contagem de Células , Linhagem da Célula , Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Osteogênese , Receptor Notch2/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteína GLI1 em Dedos de Zinco , beta Catenina/metabolismo
16.
J Bone Miner Res ; 30(9): 1585-96, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25753754

RESUMO

Articular cartilage repair remains a challenging problem. Based on a high-throughput screening and functional analysis, we found that fluocinolone acetonide (FA) in combination with transforming growth factor beta 3 (TGF-ß3) strongly potentiated chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs). In an in vivo cartilage defect model in knee joints of immunocompromised mice, transplantation of FA/TGF-ß3-treated hBMSCs could completely repair the articular surface. Analysis of the intracellular pathways revealed that FA enhanced TGF-ß3-induced phosphorylation of Smad2 and Smad3. Additionally, we performed a pathway array and found that FA activates the mTORC1/AKT pathway. Chemical inhibition of mTORC1 with rapamycin substantially suppressed FA effect, and inhibition of AKT completely repressed chondrogenesis of hBMSCs. Inhibition of glucocorticoid receptor with mifepristone also suppressed FA effect, suggesting that FA involves binding to the glucocorticoid receptor. Comparative analysis with other glucocorticoids (triamcinolone acetonide [TA] and dexamethasone [DEX]) revealed the unique ability of FA to repair articular cartilage surgical defects. Analysis of intracellular pathways showed that the mTORC1/AKT pathway and the glucocorticoid receptor was highly activated with FA and TA, but to a lesser extent with DEX. Collectively, these results show a unique ability of FA to enhance TGF-ß3-associated chondrogenesis, and suggest that the FA/TGF-ß3 combination may be used as major inducer of chondrogenesis in vitro. Additionally, FA/TGF-ß3 could be potentially applied in a clinical setting to increase the efficiency of regenerative approaches based on chondrogenic differentiation of stem cells.


Assuntos
Células da Medula Óssea/citologia , Cartilagem Articular/efeitos dos fármacos , Condrogênese/fisiologia , Fluocinolona Acetonida/química , Células-Tronco Mesenquimais/citologia , Fator de Crescimento Transformador beta3/metabolismo , Animais , Anti-Inflamatórios/química , Cartilagem Articular/metabolismo , Diferenciação Celular , Células Cultivadas , Condrogênese/efeitos dos fármacos , Dexametasona/química , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos SCID , Mifepristona/metabolismo , Receptores de Glucocorticoides/metabolismo , Regeneração , Transdução de Sinais , Triancinolona Acetonida/química
17.
Matrix Biol ; 42: 56-73, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25572963

RESUMO

It is well known that angiogenesis is linked to fibrotic processes in fibroproliferative diseases, but insights into pathophysiological processes are limited, due to lack of understanding of molecular mechanisms controlling endothelial and fibroblastic homeostasis. We demonstrate here that the matrix receptor anthrax toxin receptor 1 (ANTXR1), also known as tumor endothelial marker 8 (TEM8), is an essential component of these mechanisms. Loss of TEM8 function in mice causes reduced synthesis of endothelial basement membrane components and hyperproliferative and leaky blood vessels in skin. In addition, endothelial cell alterations in mutants are almost identical to those of endothelial cells in infantile hemangioma lesions, including activated VEGF receptor signaling in endothelial cells, increased expression of the downstream targets VEGF and CXCL12, and increased numbers of macrophages and mast cells. In contrast, loss of TEM8 in fibroblasts leads to increased rates of synthesis of fiber-forming collagens, resulting in progressive fibrosis in skin and other organs. Compromised interactions between TEM8-deficient endothelial and fibroblastic cells cause dramatic reduction in the activity of the matrix-degrading enzyme MMP2. In addition to insights into mechanisms of connective tissue homeostasis, our data provide molecular explanations for vascular and connective tissue abnormalities in GAPO syndrome, caused by loss-of-function mutations in ANTXR1. Furthermore, the loss of MMP2 activity suggests that fibrotic skin abnormalities in GAPO syndrome are, in part, the consequence of pathophysiological mechanisms underlying syndromes (NAO, Torg and Winchester) with multicentric skin nodulosis and osteolysis caused by homozygous loss-of-function mutations in MMP2.


Assuntos
Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Tecido Conjuntivo/fisiologia , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Pele/irrigação sanguínea , Alopecia/metabolismo , Alopecia/patologia , Animais , Anodontia/metabolismo , Anodontia/patologia , Linhagem Celular , Técnicas de Cocultura , Colágeno/metabolismo , Tecido Conjuntivo/embriologia , Tecido Conjuntivo/patologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Transtornos do Crescimento/metabolismo , Transtornos do Crescimento/patologia , Homeostase , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Proteínas dos Microfilamentos , Mutação , Atrofias Ópticas Hereditárias/metabolismo , Atrofias Ópticas Hereditárias/patologia , Receptores de Superfície Celular , Transdução de Sinais , Pele/embriologia , Pele/patologia
18.
Cell Rep ; 8(6): 1752-1766, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25220465

RESUMO

Cherubism is caused by mutations in SH3BP2. Studies of cherubism mice showed that tumor necrosis factor α (TNF-α)-dependent autoinflammation is a major cause of the disorder but failed to explain why human cherubism lesions are restricted to jaws and regress after puberty. We demonstrate that the inflammation in cherubism mice is MYD88 dependent and is rescued in the absence of TLR2 and TLR4. However, germ-free cherubism mice also develop inflammation. Mutant macrophages are hyperresponsive to PAMPs (pathogen-associated molecular patterns) and DAMPs (damage-associated molecular patterns) that activate Toll-like receptors (TLRs), resulting in TNF-α overproduction. Phosphorylation of SH3BP2 at Y183 is critical for the TNF-α production. Finally, SYK depletion in macrophages prevents the inflammation. These data suggest that the presence of a large amount of TLR ligands, presumably oral bacteria and DAMPs during jawbone remodeling, may cause the jaw-specific development of human cherubism lesions. Reduced levels of DAMPs after stabilization of jaw remodeling may contribute to the age-dependent regression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Querubismo/etiologia , Inflamação , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Arcada Osseodentária/diagnóstico por imagem , Fígado/patologia , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , NF-kappa B/metabolismo , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , RNA Mensageiro/metabolismo , Radiografia , Quinase Syk , Receptor 2 Toll-Like/química , Receptor 4 Toll-Like/química , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
20.
J Transl Med ; 12: 165, 2014 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-24923278

RESUMO

Progress in translational research has led to effective new treatments of a large number of diseases. Despite this progress, diseases including cancer and cardiovascular disorders still are at the top in death statistics and disorders such as osteoporosis and osteoarthritis represent an increasing disease burden in the aging population. Novel strategies in research are needed more than ever to overcome such diseases. The growing field of extracellular protein phosphorylation provides excellent opportunities to make major discoveries of disease mechanisms that can lead to novel therapies. Reversible phosphorylation/dephosphorylation of sites in the extracellular domains of matrix, cell-surface and trans-membrane proteins is emerging as a critical regulatory mechanism in health and disease. Moreover, a new concept is emerging from studies of extracellular protein phosphorylation: in cells where ATP is stored within secretory vesicles and released by exocytosis upon cell-stimulation, phosphorylation of extracellular proteins can operate as a messenger operating uniquely in signaling pathways responsible for long-term cellular adaptation. Here, we highlight new concepts that arise from this research, and discuss translation of the findings into clinical applications such as development of diagnostic disease markers and next-generation drugs.


Assuntos
Fosfoproteínas Fosfatases/metabolismo , Pesquisa Translacional Biomédica , Humanos , Fosforilação , Proteínas Quinases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA