Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
1.
Oral Dis ; 28(2): 452-468, 2022 Mar.
Article in English | MEDLINE | ID: mdl-33325564

ABSTRACT

OBJECTIVES: The project aims were to identify infectious mechanisms responsible for an extreme form of mandibular osteonecrosis and osteomyelitis in West African populations and test the hypothesis that Mycobacterium tuberculosis plays a pivotal role. MATERIALS AND METHODS: DNA was extracted from mandibular fragments of 9 of 19 patients previously included in a prospective study leading to the mycobacterial hypothesis. Amplified DNAs were used for preparing libraries suitable for next-generation sequencing. For comparison of the whole-genome sequencing data of the 9 patients with DNAs of both microbiota and human tissues, DIAMOND v0.9.26 was used to align sequencing reads to NCBI-nr database and MEGAN 6 for taxonomy binning and identification of Mycobacterium tuberculosis strains. RESULTS: The data show that mandibular bone fragments of all 9 patients not only contain Homo sapiens and Mycobacterium tuberculosis DNAs; they also contain DNAs of Plasmodium ovale wallikeri, Staphylococcus aureus, Staphylococcus hominis, and Prevotella P3-120/intermedia; as well as large numbers of DNAs from other infectious components. CONCLUSIONS: The data obtained provide direct evidence to support the conclusion that combinations of Mycobacterium tuberculosis, Plasmodium ovale wallikeri, and other oral bacteria are involved in this particular type of mandibular destruction in West African individuals of many ages.


Subject(s)
Malaria , Plasmodium ovale , Humans , Malaria/complications , Plasmodium ovale/genetics , Prospective Studies
2.
Dis Model Mech ; 14(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33563616

ABSTRACT

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.


Subject(s)
Extracellular Matrix , Vascular Endothelial Growth Factor A , Animals , Bone and Bones/metabolism , Collagen/metabolism , Extracellular Matrix/metabolism , Female , Humans , Male , Mice , Osteoblasts , Vascular Endothelial Growth Factor A/metabolism
3.
Ann Integr Mol Med ; 2(1): 75-89, 2020.
Article in English | MEDLINE | ID: mdl-32201863

ABSTRACT

The present study posits that Alzheimer's disorder is a "fast" disease. This is in sharp contrast to a view, prevailing until now, that Alzheimer's Disease (AD) is a quintessential "slow" disease that develops throughout the life as one prolonged process. According to this view, beta-amyloid (Aß) is produced and secreted solely by the beta-amyloid precursor protein (ßAPP) proteolytic/secretory pathway. As its extracellular levels increase, it triggers neurodegeneration starting relatively early in life. Damages accumulate and manifest, late in life in sporadic Alzheimer's Disease (SAD) cases, as AD symptoms. In familial AD (FAD) cases, where mutations in ßAPP gene or in presenilins increase production of either common Aß isoform or of its more toxic isoforms, neurodegeneration reaches critical threshold sooner and AD symptoms occur earlier in life, mostly in late 40s and 50s. There are currently no preventive AD therapies but if they were available, according to this viewpoint it would be largely futile to intervene late in life in case of potential SAD or at mid-age in cases of FAD because, although AD symptoms have not yet manifested, the damage has already occurred during the preceding decades. In this paradigm, to be effective, preventive therapeutic intervention should be initiated early in life. The outlook suggested by the present study is radically different. According to it, Alzheimer's disease evolves in two stages. The first stage is a slow process of intracellular beta-amyloid accumulation. It occurs via ßAPP proteolytic/secretory pathway and cellular uptake of secreted Aß common to Homo sapiens, including healthy humans, and to non-human mammals, and results neither in significant damage, nor in manifestation of the disease. The second stage occurs exclusively in humans, commences shortly before symptomatic onset of the disease, sharply accelerates the production and increases intracellular levels of Aß that is not secreted but is retained intracellularly, generates significant damages, triggers AD symptoms, and is fast. It is driven by an Aß generation pathway qualitatively and quantitatively different from ßAPP proteolytic process and entirely independent of beta-amyloid precursor protein, and results in rapid and substantial intracellular accumulation of Aß, consequent significant neurodegeneration, and symptomatic AD. In this paradigm, a preventive therapy for AD, an AD "statin", would be effective when initiated at any time prior to commencement of the second stage. Moreover, there are good reasons to believe that with a drug blocking ßAPP-independent Aß production pathway in the second stage, it would be possible not only to preempt the disease but also to stop and to reverse it even when early AD symptoms have already manifested. The present study posits a notion of AD as a Fast Disease, offers evidence for the occurrence of the AD-specific Aß production pathway, describes cellular and molecular processes constituting an engine that drives Alzheimer's disease, and explains why non-human mammals are not susceptible to AD and why only a subset of humans develop the disease. It establishes that Alzheimer's disease is preventable by therapeutic intervention initiated even late in life, details a powerful mechanism underlying the disease, suggests that Aß produced in the ßAPP-independent pathway is retained intracellularly, elaborates why neither BACE inhibition nor Aß immunotherapy are effective in treatment of AD and why intracellularly retained beta-amyloid could be the primary agent of neuronal death in Alzheimer's disease, necessitates generation of a novel animal AD model capable of producing Aß via ßAPP-independent pathway, proposes therapeutic targets profoundly different from previously pursued components of the ßAPP proteolytic pathway, and provides conceptual rationale for design of drugs that could be used not only preemptively but also for treatment and reversal of the early stages of the disease.

4.
Ann Integr Mol Med ; 1(1): 61-74, 2019.
Article in English | MEDLINE | ID: mdl-31858090

ABSTRACT

The present study defines RNA-dependent amplification of ßAPP mRNA as a molecular basis of beta-amyloid overproduction in Alzheimer's disease. In this process, ßAPP mRNA serves as a template for RNA-dependent RNA polymerase, RdRp complex. The resulting antisense RNA self-primes its extension utilizing two complementary elements: 3'-terminal and internal, located within an antisense segment corresponding to the coding portion of ßAPP mRNA. The extension produces 3'-terminal fragment of ßAPP mRNA, a part of a hairpin-structured antisense/sense RNA molecule. Cleavage at the 3' end of the hairpin loop produces RNA end product encoding a C-terminal fragment of ßAPP. Since each conventional ßAPP mRNA can be used repeatedly as a template, the process constitutes an asymmetric mRNA amplification. The 5'-most translation initiation codon of the amplified mRNA is the AUG preceding immediately and in-frame the Aß-coding segment. Translation from this codon overproduces Aß independently of ßAPP. Such process can occur in humans but not in mice and other animals where segments of ßAPP antisense RNA required for self-priming have little, if any, complementarity. This explains why Alzheimer's disease occurs exclusively in humans and implies that ßAPP mRNA amplification is requisite in AD. In AD, therefore, there are two pathways of beta-amyloid production: ßAPP proteolytic pathway and ßAPP mRNA amplification pathway independent of ßAPP and insensitive to beta-secretase inhibition. This implies that in healthy humans, where only the proteolytic pathway is in operation, Aß production should be suppressed by the BACE inhibition, and indeed it is. However, since ßAPP-independent pathway operating in AD is by far the predominant one, BACE inhibition has no effect in Alzheimer's disease. It appears that, physiologically, the extent of beta-amyloid overproduction sufficient to trigger amyloid cascade culminating in AD requires asymmetric RNA-dependent amplification of ßAPP mRNA and cannot be reached without it. In turn, the occurrence of mRNA amplification process depends on the activation of inducible components of RdRp complex by certain stresses, for example the ER stress in case of amplification of mRNA encoding extracellular matrix proteins. In case of Alzheimer's disease, such an induction appears to be triggered by stresses associated with mitochondrial dysfunction, a phenomenon closely linked to AD. The cause-and-effect relationships between mitochondrial dysfunction and AD appear to be very different in familial, FAD, and sporadic, SAD cases. In FAD, increased levels or more toxic species of Aß resulting from the abnormal proteolysis of ßAPP trigger mitochondrial dysfunction, activate mRNA amplification and increase the production of Aß, reinforcing the cycle. Thus in FAD, mitochondrial dysfunction is an intrinsic component of the amyloid cascade. The reverse sequence is true in SAD where aging-related mitochondrial dysfunction activates amplification of ßAPP mRNA and enhances the production of Aß. This causes further mitochondrial dysfunction, the cycle repeats and degeneration increases. Thus in SAD, the initial mitochondrial dysfunction arises prior to the disease, independently of and upstream from the increased Aß production, i.e. in SAD, mitochondrial pathology hierarchically supersedes Aß pathology. This is the primary reason for the formulation of the Mitochondrial Cascade Hypothesis. But even in terms of the MCH, the core of the disease is the amyloid cascade as defined in the amyloid cascade hypothesis, ACH. The role of mitochondrial dysfunction in relation to this core is causative in SAD and auxiliary in FAD. In FAD, the initial increase in the production of Aß is mutations-based and occurs relatively early in life, whereas in SAD it is coerced by an aging-contingent component, but both lead to mechanistically identical self-perpetuating mutual Aß/mitochondrial dysfunction feedback cycles, an engine that drives, via RNA-dependent ßAPP mRNA amplification, overproduction of beta-amyloid and, consequently, AD; hence drastic difference in the age of onset, yet profound pathological and symptomatic similarity in the progression, of familial and sporadic forms of Alzheimer's disease. Interestingly, the recent findings that mitochondrial microprotein PIGBOS interacts with the ER in mitigating the unfolded protein response indicate a possible connection between mitochondrial dysfunction and ER stress, implicated in activation of RNA-dependent mRNA amplification pathway. The possible involvement of mitochondrial dysfunction in ßAPP mRNA amplification makes it a promising therapeutic target. Recent successes in mitigating, and even reversing, Aß-induced metabolic defects with anti-diabetes drug metformin are encouraging in this respect.

5.
Ann Integr Mol Med ; 1(1): 23-47, 2019.
Article in English | MEDLINE | ID: mdl-31656957

ABSTRACT

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.

6.
Ann Integr Mol Med ; 1(1): 48-60, 2019.
Article in English | MEDLINE | ID: mdl-31663081

ABSTRACT

De novo production of RNA on RNA template, a process known as RNA-dependent RNA synthesis, RdRs, and the enzymatic activity conducting it, RNA-dependent RNA polymerase, RdRp, were initially considered to be exclusively virus-specific. Eventually, however, the occurrence of RdRs and the ubiquitous presence of conventional RdRp were demonstrated in numerous eukaryotic organisms. The evidence that the enzymatic machinery capable of RdRs is present in mammalian cells was derived from studies of viruses, such as hepatitis delta virus, HDV, that do not encode RdRp yet undergo a robust RNA replication once inside the mammalian host; thus firmly establishing its occurrence and functionality. Moreover, 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, given 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 RNAs 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. Recent detection of the major model-predicted identifiers, chimeric RNA intermediates containing both sense and antisense RNA strands covalently joined in a rigorously predicted and uniquely defined manner, as well as the identification of a putative chimeric RNA end product of this process, validated the proposed model. The results corroborating mammalian RNA-dependent mRNA amplification were obtained in vivo with cells undergoing terminal erythroid differentiation and programmed for only a short survival span. This raises a question of whether mammalian RNA-dependent mRNA amplification is a specialized occurrence limited to extreme circumstances of terminal differentiation or a general physiological phenomenon. The present study addresses this question by testing for the occurrence of RNA-dependent amplification of mRNA encoding extracellular matrix proteins abundantly produced throughout the tissue and organ development and homeostasis, an exceptionally revealing indicator of the range and scope of this phenomenon. We report here the detection of major identifiers of RNA-dependent amplification of mRNA encoding α1, ß1, and γ1 chains of laminin in mouse tissues producing large quantities of extracellular matrix proteins. The results obtained warrant reinterpretation of the mechanisms involved in ubiquitous and abundant production and deposition of extracellular matrix proteins, confirm the occurrence of mammalian RNA-dependent mRNA amplification as a new mode of genomic protein-encoding information transfer, and establish it as a general physiological phenomenon.

7.
J Bone Miner Res ; 34(11): 2117-2132, 2019 11.
Article in English | MEDLINE | ID: mdl-31269275

ABSTRACT

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.


Subject(s)
Bone Development , Bone Marrow Cells/metabolism , Bone and Bones/blood supply , Endothelial Cells/metabolism , Sex Characteristics , Vascular Endothelial Growth Factor A/metabolism , Animals , Bone and Bones/metabolism , Female , Male , Mice , Mice, Knockout , Vascular Endothelial Growth Factor A/genetics
9.
J Dent Educ ; 82(11): 1171-1177, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30385683

ABSTRACT

For many years, Harvard School of Dental Medicine (HSDM) has had a research requirement for predoctoral students, but a recent curriculum assessment prompted a re-examination of that requirement and how it is implemented. The aim of this retrospective study was to assess the students' perspectives on research in a predoctoral dental program in which a research experience was mandatory for graduation. Data that had been collected in graduating student surveys from 2008 to 2017 were analyzed to gauge students' perceptions of the research program in the previous curriculum (New Pathways) and to seek insights to improve the next generation of this requirement in the Pathways curriculum. In the results, 74% of the students reported their research experience had a positive impact on their dental education. Half of the students (49%) indicated they would have pursued research even if it had not been a graduation requirement, while 37% were uncertain. A large majority (82%) said they would remain involved in research during their dental career. The majority of these HSDM students had contributed to scholarship with presentations and published results, planning of projects, or preparation of manuscripts. These results show that the research requirement has had a positive effect on students' perspectives on research as a part of their careers.


Subject(s)
Attitude , Dental Research , Education, Dental/standards , Students, Dental/psychology , Curriculum , Retrospective Studies , Self Report , Time Factors
10.
Proc Natl Acad Sci U S A ; 115(36): 9008-9013, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30126991

ABSTRACT

Ligaments serve as compliant connectors between hard tissues. In that role, they function under various load regimes and directions. The 3D structure of ligaments is considered to form as a uniform entity that changes due to function. The periodontal ligament (PDL) connects the tooth to the bone and sustains different types of loads in various directions. Using the PDL as a model, employing a fabricated motorized setup in a microCT, we demonstrate that the fibrous network structure within the PDL is not uniform, even before the tooth becomes functional. Utilizing morphological automated segmentation methods, directionality analysis, as well as second harmonic generation imaging, we find high correlation between blood vessel distribution and fiber density. We also show a structural feature in a form of a dense collar around the neck of the tooth as well as a preferred direction of the fibrous network. Finally, we show that the PDL develops as a nonuniform structure, with an architecture designed to sustain specific types of load in designated areas. Based on these findings, we propose that ligaments in general should be regarded as nonuniform entities, structured already at developmental stages for optimal functioning under variable load regimes.


Subject(s)
Periodontal Ligament/diagnostic imaging , Tooth/diagnostic imaging , X-Ray Microtomography , Animals , Mice , Mice, Transgenic , Periodontal Ligament/blood supply , Periodontal Ligament/metabolism , Tooth/blood supply , Tooth/metabolism
11.
Sci Rep ; 7(1): 13027, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29026147

ABSTRACT

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.


Subject(s)
Cartilage, Articular/embryology , Cartilage, Articular/pathology , Osteoarthritis/metabolism , Vascular Endothelial Growth Factor A/metabolism , Administration, Oral , Animals , Antibodies/pharmacology , Bone Development , Cell Differentiation , Cell Lineage , Chondrocytes/metabolism , Collagen Type II/metabolism , Disease Progression , Endothelium/metabolism , Female , Gene Deletion , Gene Expression Regulation , Growth Plate/metabolism , Growth Plate/pathology , Integrases/metabolism , Knee Joint/pathology , Mice, Inbred C57BL , Neovascularization, Physiologic , Osteoarthritis/pathology , Osteogenesis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor A/deficiency , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Biotechnol J ; 12(12)2017 Dec.
Article in English | MEDLINE | ID: mdl-28762648

ABSTRACT

Neural crest stem cells (NCSCs) are a transient and multipotent cell population giving rise to various cell types with clinical importance. Isolation of human NCSCs is extremely challenging that limits our knowledge about neural crest development and application. Here, a defined protocol to efficiently direct human embryonic stem cells (hESCs) to NCSCs and multiple neural crest lineages is presented. A unique combination of small molecule inhibitors and growth factors is employed to generate NCSCs from hESCs through a neuroectoderm stage. The self-renewal and multipotent capacities of hESC-derived NCSCs are assessed subsequently. In the feeder-free system, hESC-derived NCSCs (P75+ /HNK1+ /AP2α+ /PAX6- ) in high purity are efficiently generated following neuroectodermal restriction. They can be propagated and differentiated toward multiple neural crest lineages in vitro, such as functional peripheral neurons (ß-tubulin III+ /peripherin+ ), mesenchymal stem cells (CD73+ CD90+ CD105+ ), and corneal keratocytes (keratocan+ ). The in vivo developmental potential of hESC-derived NCSCs is confirmed using zebrafish embryos. This report is the first demonstration of efficient differentiation of hESCs into corneal keratocytes as a monolayer in a feeder-free system. Considering the high efficacy of NCSC generation, this new method will be a useful tool for future clinical organ repair and regeneration, such as peripheral nerve regeneration and corneal repair.


Subject(s)
Corneal Keratocytes/cytology , Human Embryonic Stem Cells/cytology , Neural Crest/cytology , Neural Stem Cells/cytology , Neurogenesis/physiology , Animals , Cell Culture Techniques/methods , Cells, Cultured , Embryo, Nonmammalian , Flow Cytometry , Humans , Neurons/cytology , Zebrafish
13.
Matrix Biol ; 62: 105-114, 2017 10.
Article in English | MEDLINE | ID: mdl-28011198

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/genetics , Collagen Type I/metabolism , Fibroblasts/metabolism , Fibronectins/metabolism , Receptors, Peptide/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Cells, Cultured , Collagen Type I, alpha 1 Chain , Connective Tissue Growth Factor/metabolism , Extracellular Matrix/metabolism , Gene Knockout Techniques , Homeostasis , Mice , Microfilament Proteins , Receptors, Cell Surface , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
14.
J Orthop Res ; 35(7): 1461-1469, 2017 07.
Article in English | MEDLINE | ID: mdl-27658810

ABSTRACT

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.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Fracture Healing/drug effects , Osteogenesis/drug effects , Vascular Endothelial Growth Factor Receptor-1/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Protein 2/therapeutic use , Drug Evaluation, Preclinical , Fractures, Bone/drug therapy , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , Neovascularization, Physiologic/drug effects , Osteoclasts/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-1/therapeutic use
15.
Dev Dyn ; 246(4): 227-234, 2017 04.
Article in English | MEDLINE | ID: mdl-27750398

ABSTRACT

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.


Subject(s)
Bone Development , Bone Regeneration , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factors/physiology , Animals , Bone Diseases, Developmental/congenital , Bone Remodeling , Bone and Bones , Humans
16.
Int J Exp Pathol ; 97(4): 296-302, 2016 08.
Article in English | MEDLINE | ID: mdl-27581728

ABSTRACT

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.


Subject(s)
Bone Development/physiology , Connective Tissue/growth & development , Hemangioma/metabolism , Homeostasis/physiology , Animals , Hemangioma/pathology , Humans , Microfilament Proteins , Neoplasm Proteins/physiology , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
Bone ; 91: 30-8, 2016 10.
Article in English | MEDLINE | ID: mdl-27353702

ABSTRACT

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.


Subject(s)
Bone Regeneration , Vascular Endothelial Growth Factor A/metabolism , Wound Healing , Animals , Humans , Osteoblasts/metabolism , Osteogenesis , Osteogenesis, Distraction
18.
J Bone Miner Res ; 31(5): 911-24, 2016 05.
Article in English | MEDLINE | ID: mdl-27163679

ABSTRACT

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.


Subject(s)
Osteoarthritis/drug therapy , Pain/drug therapy , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Female , Humans , Male , Osteoarthritis/metabolism , Pain/metabolism , Vascular Endothelial Growth Factor A/metabolism
19.
PLoS One ; 11(3): e0152833, 2016.
Article in English | MEDLINE | ID: mdl-27031698

ABSTRACT

Vascular abnormalities in the eye are the leading cause of many forms of inherited and acquired human blindness. Loss-of-function mutations in the Wnt-binding co-receptor LRP5 leads to aberrant ocular vascularization and loss of vision in genetic disorders such as osteoporosis-pseudoglioma syndrome. The canonical Wnt-ß-catenin pathway is known to regulate retinal vascular development. However, it is unclear what precise role LPR5 plays in this process. Here, we show that loss of LRP5 function in mice causes retinal hypovascularization during development as well as retinal neovascularization in adulthood with disorganized and leaky vessels. Using a highly specific Flk1-CreBreier line for vascular endothelial cells, together with several genetic models, we demonstrate that loss of endothelium-derived LRP5 recapitulates the retinal vascular defects in Lrp5-/- mice. In addition, restoring LRP5 function only in endothelial cells in Lrp5-/- mice rescues their retinal vascular abnormalities. Furthermore, we show that retinal vascularization is regulated by LRP5 in a dosage dependent manner and does not depend on LRP6. Our study provides the first direct evidence that endothelium-derived LRP5 is both necessary and sufficient to mediate its critical role in the development and maintenance of retinal vasculature.


Subject(s)
Endothelial Cells/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Retina/growth & development , Retinal Vessels/growth & development , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Gene Deletion , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Mice , Receptor, TIE-2/metabolism , Retina/metabolism , Retinal Neovascularization/genetics , Retinal Neovascularization/metabolism , Retinal Vessels/abnormalities , Signal Transduction
20.
Matrix Biol ; 52-54: 127-140, 2016.
Article in English | MEDLINE | ID: mdl-26899202

ABSTRACT

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.


Subject(s)
Mandible/growth & development , Neural Crest/cytology , Skull/growth & development , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Animals , Animals, Newborn , Calcification, Physiologic , Cell Proliferation , Cells, Cultured , Collagen Type II/metabolism , Gene Knockdown Techniques , Mandible/metabolism , Mice , Skull/metabolism , Sp7 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...