Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Pediatr Neurol ; 143: 34-43, 2023 06.
Article in English | MEDLINE | ID: mdl-36996759

ABSTRACT

BACKGROUND: Neurofibromatosis type 1 (NF1) is a genetic neurocutaneous disorder commonly associated with motor and cognitive symptoms that greatly impact quality of life. Transcranial magnetic stimulation (TMS) can quantify motor cortex physiology, reflecting the basis for impaired motor function as well as, possibly, clues for mechanisms of effective treatment. We hypothesized that children with NF1 have impaired motor function and altered motor cortex physiology compared to typically developing (TD) control children and children with attention-deficit/hyperactivity disorder (ADHD). METHODS: Children aged 8-17 years with NF1 (n = 21) were compared to children aged 8-12 years with ADHD (n = 59) and TD controls (n = 88). Motor development was assessed using the Physical and Neurological Examination for Subtle Signs (PANESS) scale. The balance of inhibition and excitation in motor cortex was assessed using the TMS measures short-interval cortical inhibition (SICI) and intracortical facilitation (ICF). Measures were compared by diagnosis and tested using bivariate correlations and regression for association with clinical characteristics. RESULTS: In NF1, ADHD severity scores were intermediate between the ADHD and TD cohorts, but total PANESS scores were markedly elevated (worse) compared to both (P < 0.001). Motor cortex ICF (excitatory) was significantly lower in NF1 than in TD and ADHD (P < 0.001), but SICI (inhibitory) did not differ. However, in NF1, better PANESS scores correlated with lower SICI ratios (more inhibition; ρ = 0.62, P = 0.003) and lower ICF ratios (less excitation; ρ = 0.38, P = 0.06). CONCLUSIONS: TMS-evoked SICI and ICF may reflect processes underlying abnormal motor function in children with NF1.


Subject(s)
Neural Inhibition , Neurofibromatosis 1 , Child , Humans , Adolescent , Neural Inhibition/physiology , Neurofibromatosis 1/complications , Quality of Life , Evoked Potentials, Motor/physiology , Electromyography , Transcranial Magnetic Stimulation
2.
Acta Neuropathol Commun ; 6(1): 127, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30470263

ABSTRACT

Normal Schwann cells (SCs) are quiescent in adult nerves, when ATP is released from the nerve in an activity dependent manner. We find that suppressing nerve activity in adult nerves causes SC to enter the cell cycle. In vitro, ATP activates the SC G-protein coupled receptor (GPCR) P2Y2. Downstream of P2Y2, ß-arrestin-mediated signaling results in PP2-mediated de-phosphorylation of AKT, and PP2 activity is required for SC growth suppression. NF1 deficient SC show reduced growth suppression by ATP, and are resistant to the effects of ß-arrestin-mediated signaling, including PP2-mediated de-phosphorylation of AKT. In patients with the disorder Neurofibromatosis type 1, NF1 mutant SCs proliferate and form SC tumors called neurofibromas. Elevating ATP levels in vivo reduced neurofibroma cell proliferation. Thus, the low proliferation characteristic of differentiated adult peripheral nerve may require ongoing, nerve activity-dependent, ATP. Additionally, we identify a mechanism through which NF1 SCs may evade growth suppression in nerve tumors.


Subject(s)
Adenosine Triphosphate/metabolism , Arrestin/metabolism , Neurofibromin 1/deficiency , Neuroglia/metabolism , Protein Phosphatase 2/metabolism , Sciatic Nerve/cytology , Animals , Bupivacaine/pharmacology , Calcium/metabolism , Cells, Cultured , Embryo, Mammalian , Ganglia, Spinal/cytology , Humans , Hydroxides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurofibromin 1/genetics , Neuroglia/drug effects , Neurons/drug effects , Neurons/metabolism , Pain Measurement , Sciatic Neuropathy , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
3.
N Engl J Med ; 375(26): 2550-2560, 2016 12 29.
Article in English | MEDLINE | ID: mdl-28029918

ABSTRACT

BACKGROUND: Effective medical therapies are lacking for the treatment of neurofibromatosis type 1-related plexiform neurofibromas, which are characterized by elevated RAS-mitogen-activated protein kinase (MAPK) signaling. METHODS: We conducted a phase 1 trial of selumetinib (AZD6244 or ARRY-142886), an oral selective inhibitor of MAPK kinase (MEK) 1 and 2, in children who had neurofibromatosis type 1 and inoperable plexiform neurofibromas to determine the maximum tolerated dose and to evaluate plasma pharmacokinetics. Selumetinib was administered twice daily at a dose of 20 to 30 mg per square meter of body-surface area on a continuous dosing schedule (in 28-day cycles). We also tested selumetinib using a mouse model of neurofibromatosis type 1-related neurofibroma. Response to treatment (i.e., an increase or decrease from baseline in the volume of plexiform neurofibromas) was monitored by using volumetric magnetic resonance imaging analysis to measure the change in size of the plexiform neurofibroma. RESULTS: A total of 24 children (median age, 10.9 years; range, 3.0 to 18.5) with a median tumor volume of 1205 ml (range, 29 to 8744) received selumetinib. Patients were able to receive selumetinib on a long-term basis; the median number of cycles was 30 (range, 6 to 56). The maximum tolerated dose was 25 mg per square meter (approximately 60% of the recommended adult dose). The most common toxic effects associated with selumetinib included acneiform rash, gastrointestinal effects, and asymptomatic creatine kinase elevation. The results of pharmacokinetic evaluations of selumetinib among the children in this trial were similar to those published for adults. Treatment with selumetinib resulted in confirmed partial responses (tumor volume decreases from baseline of ≥20%) in 17 of the 24 children (71%) and decreases from baseline in neurofibroma volume in 12 of 18 mice (67%). Disease progression (tumor volume increase from baseline of ≥20%) has not been observed to date. Anecdotal evidence of decreases in tumor-related pain, disfigurement, and functional impairment was observed. CONCLUSIONS: Our early-phase data suggested that children with neurofibromatosis type 1 and inoperable plexiform neurofibromas benefited from long-term dose-adjusted treatment with selumetinib without having excess toxic effects. (Funded by the National Institutes of Health and others; ClinicalTrials.gov number, NCT01362803 .).


Subject(s)
Benzimidazoles/administration & dosage , Benzimidazoles/pharmacokinetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neurofibroma, Plexiform/drug therapy , Neurofibromatosis 1/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Adolescent , Animals , Benzimidazoles/adverse effects , Child , Child, Preschool , Disease Models, Animal , Disease Progression , Female , Humans , Magnetic Resonance Imaging , Male , Mice , Neurofibroma, Plexiform/diagnostic imaging , Protein Kinase Inhibitors/adverse effects
4.
Dev Biol ; 405(1): 96-107, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26141957

ABSTRACT

The sequence of events that leads to the formation of a functionally graded enthesis is not clearly defined. The current study demonstrates that clonal expansion of Gdf5 progenitors contributes to linear growth of the enthesis. Prior to mineralization, Col1+ cells in the enthesis appose Col2+ cells of the underlying primary cartilage. At the onset of enthesis mineralization, cells at the base of the enthesis express alkaline phosphatase, Indian hedgehog, and ColX as they mineralize. The mineralization front then extends towards the tendon midsubstance as cells above the front become encapsulated in mineralized fibrocartilage over time. The hedgehog (Hh) pathway regulates this process, as Hh-responsive Gli1+ cells within the developing enthesis mature from unmineralized to mineralized fibrochondrocytes in response to activated signaling. Hh signaling is required for mineralization, as tissue-specific deletion of its obligate transducer Smoothened in the developing tendon and enthesis cells leads to significant reductions in the apposition of mineralized fibrocartilage. Together, these findings provide a spatiotemporal map of events - from expansion of the embryonic progenitor pool to synthesis of the collagen template and finally mineralization of this template - that leads to the formation of the mature zonal enthesis. These results can inform future tendon-to-bone repair strategies to create a mechanically functional enthesis in which tendon collagen fibers are anchored to bone through mineralized fibrocartilage.


Subject(s)
Fibrocartilage/cytology , Growth Differentiation Factor 5/metabolism , Hedgehog Proteins/metabolism , Minerals/metabolism , Signal Transduction , Stem Cells/cytology , Animals , Bone Marrow/pathology , Bone Resorption/pathology , Bone Resorption/physiopathology , Bone and Bones/physiology , Calcification, Physiologic , Cell Differentiation , Chondrocytes/metabolism , Clone Cells , Collagen/metabolism , Epiphyses/pathology , Integrases/metabolism , Kruppel-Like Transcription Factors/metabolism , Mice , Models, Biological , Osteoclasts/metabolism , Patella/physiology , Staining and Labeling , Stem Cells/metabolism , Tendons/physiology , Zinc Finger Protein GLI1
5.
J Orthop Res ; 33(8): 1142-51, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25807894

ABSTRACT

Restoring the native structure of the tendon enthesis, where collagen fibers of the midsubstance are integrated within a fibrocartilaginous structure, is problematic following injury. As current surgical methods fail to restore this region adequately, engineers, biologists, and clinicians are working to understand how this structure forms as a prerequisite to improving repair outcomes. We recently reported on the role of Indian hedgehog (Ihh), a novel enthesis marker, in regulating early postnatal enthesis formation. Here, we investigate how inactivating the Hh pathway in tendon cells affects adult (12-week) murine patellar tendon (PT) enthesis mechanics, fibrocartilage morphology, and collagen fiber organization. We show that ablating Hh signaling resulted in greater than 100% increased failure insertion strain (0.10 v. 0.05 mm/mm, p<0.01) as well as sub-failure biomechanical deficiencies. Although collagen fiber orientation appears overtly normal in the midsubstance, ablating Hh signaling reduces mineralized fibrocartilage by 32%, leading to less collagen embedded within mineralized tissue. Ablating Hh signaling also caused collagen fibers to coalesce at the insertion, which may explain in part the increased strains. These results indicate that Ihh signaling plays a critical role in the mineralization process of fibrocartilaginous entheses and may be a novel therapeutic to promote tendon-to-bone healing.


Subject(s)
Connective Tissue/physiology , Hedgehog Proteins/physiology , Patella/physiology , Signal Transduction/physiology , Tendons/physiology , Animals , Biomechanical Phenomena , Male , Mice , Mice, Knockout , Tendons/cytology
6.
PLoS One ; 8(6): e65411, 2013.
Article in English | MEDLINE | ID: mdl-23762363

ABSTRACT

Tendons are typically composed of two histologically different regions: the midsubstance and insertion site. We previously showed that Gli1, a downstream effector of the hedgehog (Hh) signaling pathway, is expressed only in the insertion site of the mouse patellar tendon during its differentiation. To test for a functional role of Hh signaling, we targeted the Smoothened (Smo) gene in vivo using a Cre/Lox system. Constitutive activation of the Hh pathway in the mid-substance caused molecular markers of the insertion site, e.g. type II collagen, to be ectopically expressed or up-regulated in the midsubstance. This was confirmed using a novel organ culture method in vitro. Conversely, when Smo was excised in the scleraxis-positive cell population, the development of the fibrocartilaginous insertion site was affected. Whole transcriptome analysis revealed that the expression of genes involved in chondrogenesis and mineralization was down-regulated in the insertion site, and expression of insertion site markers was decreased. Biomechanical testing of murine adult patellar tendon, which developed in the absence of Hh signaling, showed impairment of tendon structural properties (lower linear stiffness and greater displacement) and material properties (greater strain), although the linear modulus of the mutant group was not significantly lower than controls. These studies provide new insights into the role of Hh signaling during tendon development.


Subject(s)
Cell Differentiation , Cytoskeletal Proteins/physiology , Fibrocartilage/cytology , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Muscle Proteins/physiology , Patellar Ligament/cytology , Animals , Biomarkers/metabolism , Blotting, Western , Cell Proliferation , Female , Fibrocartilage/metabolism , Gene Expression Profiling , Hedgehog Proteins/genetics , Immunoenzyme Techniques , Integrases , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Organ Culture Techniques , Patellar Ligament/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Zinc Finger Protein GLI1
7.
PLoS One ; 8(3): e59944, 2013.
Article in English | MEDLINE | ID: mdl-23555841

ABSTRACT

The origin of cells that contribute to tendon healing, specifically extrinsic epitenon/paratenon cells vs. internal tendon fibroblasts, is still debated. The purpose of this study is to determine the location and phenotype of cells that contribute to healing of a central patellar tendon defect injury in the mouse. Normal adult patellar tendon consists of scleraxis-expressing (Scx) tendon fibroblasts situated among aligned collagen fibrils. The tendon body is surrounded by paratenon, which consists of a thin layer of cells that do not express Scx and collagen fibers oriented circumferentially around the tendon. At 3 days following injury, the paratenon thickens as cells within the paratenon proliferate and begin producing tenascin-C and fibromodulin. These cells migrate toward the defect site and express scleraxis and smooth muscle actin alpha by day 7. The thickened paratenon tissue eventually bridges the tendon defect by day 14. Similarly, cells within the periphery of the adjacent tendon struts express these markers and become disorganized. Cells within the defect region show increased expression of fibrillar collagens (Col1a1 and Col3a1) but decreased expression of tenogenic transcription factors (scleraxis and mohawk homeobox) and collagen assembly genes (fibromodulin and decorin). By contrast, early growth response 1 and 2 are upregulated in these tissues along with tenascin-C. These results suggest that paratenon cells, which normally do not express Scx, respond to injury by turning on Scx and assembling matrix to bridge the defect. Future studies are needed to determine the signaling pathways that drive these cells and whether they are capable of producing a functional tendon matrix. Understanding this process may guide tissue engineering strategies in the future by stimulating these cells to improve tendon repair.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Gene Expression Regulation , Patellar Ligament/injuries , Patellar Ligament/metabolism , Tendon Injuries/metabolism , Actins/metabolism , Animals , Cell Movement , Collagen/metabolism , Extracellular Matrix Proteins/biosynthesis , Fibromodulin , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Mice , Muscle, Smooth/metabolism , Phenotype , Principal Component Analysis , Proteoglycans/biosynthesis , Tenascin/biosynthesis , Time Factors , Wound Healing/genetics
8.
Tissue Eng Part A ; 18(5-6): 598-608, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21939397

ABSTRACT

Tendon injuries are common clinical problems and are difficult to treat. In particular, the tendon-to-bone insertion site, once damaged, does not regenerate its complex zonal arrangement. A potential treatment for tendon injuries is to replace injured tendons with bioengineered tendons. However, the bioengineering of tendon will require a detailed understanding of the normal development of tendon, which is currently lacking. Here, we use the mouse patellar tendon as a model to describe the spatial and temporal pattern of expression of molecular markers for tendon differentiation from late fetal life to 2 weeks after birth. We found that collagen I, fibromodulin, and tenomodulin were expressed throughout the tendon, whereas tenascin-C, biglycan, and cartilage oligomeric protein were concentrated in the insertion site during this period. We also identified signaling pathways that are activated both throughout the developing tendon, for example, transforming growth factor beta and bone morphogenetic protein, and specifically in the insertion site, for example, hedgehog pathway. Using a mouse line expressing green fluorescent protein in all tenocytes, we also found that tenocyte cell proliferation occurs at highest levels during late fetal life, and declines to very low levels by 2 weeks after birth. These data will allow both the functional analysis of specific signaling pathways in tenocyte development and their application to tissue-engineering studies in vitro.


Subject(s)
Antigens, Differentiation/biosynthesis , Cell Proliferation , Patellar Ligament , Signal Transduction/physiology , Animals , Hedgehog Proteins/metabolism , Matrix Metalloproteinases, Secreted/metabolism , Mice , Mice, Transgenic , Patellar Ligament/cytology , Patellar Ligament/embryology , Patellar Ligament/growth & development , Tendon Injuries/metabolism , Tendon Injuries/therapy , Tissue Engineering , Transforming Growth Factor beta/metabolism
9.
J Orthop Res ; 30(1): 28-36, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21698662

ABSTRACT

Tendon injuries are major orthopedic problems that worsen as the population ages. Type-I (Col1) and type-II (Col2) collagens play important roles in tendon midsubstance and tendon-to-bone insertion healing, respectively. Using double transgenic mice, this study aims to spatiotemporally monitor Col1 and Col2 gene expression, histology, and biomechanics up to 8 weeks following a full-length patellar tendon injury. Gene expression and histology were analyzed weekly for up to 5 weeks while mechanical properties were measured at 1, 2, 5, and 8 weeks. At week 1, the healing region displayed loose granulation tissue with little Col1 expression. Col1 expression peaked at 2 weeks, but the ECM was highly disorganized and hypercellular. By 3 weeks, Col1 expression had reduced and by 5 weeks, the ECM was generally aligned along the tendon axis. Col2 expression was not seen in the healing midsubstance or insertion at any time point. The biomechanics of the healing tissue was inadequate at all time points, achieving ultimate loads and stiffnesses of 48% and 63% of normal values by 8 weeks. Future studies will further characterize the cells within the healing midsubstance and insertion using tenogenic markers and compare these results to those of tendon cells during normal development.


Subject(s)
Collagen Type II/genetics , Collagen Type I/genetics , Knee Injuries , Patellar Ligament , Tendon Injuries , Animals , Biomechanical Phenomena/physiology , Disease Models, Animal , Extracellular Matrix/physiology , Knee Injuries/genetics , Knee Injuries/pathology , Knee Injuries/physiopathology , Mice , Mice, Transgenic , Patellar Ligament/injuries , Patellar Ligament/physiopathology , Patellar Ligament/surgery , Tendon Injuries/genetics , Tendon Injuries/pathology , Tendon Injuries/physiopathology , Weight-Bearing/physiology , Wound Healing/physiology
10.
Tissue Eng Part B Rev ; 17(3): 165-76, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21314435

ABSTRACT

Tendons connect muscles to bones, and serve as the transmitters of force that allow all the movements of the body. Tenocytes are the basic cellular units of tendons, and produce the collagens that form the hierarchical fiber system of the tendon. Tendon injuries are common, and difficult to repair, particularly in the case of the insertion of tendon into bone. Successful attempts at cell-based repair therapies will require an understanding of the normal development of tendon tissues, including their differentiated regions such as the fibrous mid-section and fibrocartilaginous insertion site. Many genes are known to be involved in the formation of tendon. However, their functional roles in tendon development have not been fully characterized. Tissue engineers have attempted to generate functional tendon tissue in vitro. However, a lack of knowledge of normal tendon development has hampered these efforts. Here we review studies focusing on the developmental mechanisms of tendon development, and discuss the potential applications of a molecular understanding of tendon development to the treatment of tendon injuries.


Subject(s)
Models, Biological , Tendon Injuries/physiopathology , Tendon Injuries/surgery , Tendons/physiopathology , Tissue Engineering , Animals , Humans , Tendon Injuries/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...