44 research outputs found

    Human Muscle Progenitor Cells Overexpressing Neurotrophic Factors Improve Neuronal Regeneration in a Sciatic Nerve Injury Mouse Model

    Get PDF
    The peripheral nervous system has an intrinsic ability to regenerate after injury. However, this process is slow, incomplete, and often accompanied by disturbing motor and sensory consequences. Sciatic nerve injury (SNI), which is the most common model for studying peripheral nerve injury, is characterized by damage to both motor and sensory fibers. The main goal of this study is to examine the feasibility of administration of human muscle progenitor cells (hMPCs) overexpressing neurotrophic factor (NTF) genes, known to protect peripheral neurons and enhance axon regeneration and functional recovery, to ameliorate motoric and sensory deficits in SNI mouse model. To this end, hMPCs were isolated from a human muscle biopsy, and manipulated to ectopically express brain-derived neurotrophic factor (BDNF), glial-cell-line-derived neurotrophic factor (GDNF), vascular endothelial growth factor (VEGF), and insulin-like growth factor (IGF-1). These hMPC-NTF were transplanted into the gastrocnemius muscle of mice after SNI, and motor and sensory functions of the mice were assessed using the CatWalk XT system and the hot plate test. ELISA analysis showed that genetically manipulated hMPC-NTF express significant amounts of BDNF, GDNF, VEGF, or IGF-1. Transplantation of 3 × 106 hMPC-NTF was shown to improve motor function and gait pattern in mice following SNI surgery, as indicated by the CatWalk XT system 7 days post-surgery. Moreover, using the hot-plate test, performed 6 days after surgery, the treated mice showed less sensory deficits, indicating a palliative effect of the treatment. ELISA analysis following transplantation demonstrated increased NTF expression levels in the gastrocnemius muscle of the treated mice, reinforcing the hypothesis that the observed positive effect was due to the transplantation of the genetically manipulated hMPC-NTF. These results show that genetically modified hMPC can alleviate both motoric and sensory deficits of SNI. The use of hMPC-NTF demonstrates the feasibility of a treatment paradigm, which may lead to rapid, high-quality healing of damaged peripheral nerves due to administration of hMPC. Our approach suggests a possible clinical application for the treatment of peripheral nerve injury

    Expected Performance of the ATLAS Experiment - Detector, Trigger and Physics

    Get PDF
    A detailed study is presented of the expected performance of the ATLAS detector. The reconstruction of tracks, leptons, photons, missing energy and jets is investigated, together with the performance of b-tagging and the trigger. The physics potential for a variety of interesting physics processes, within the Standard Model and beyond, is examined. The study comprises a series of notes based on simulations of the detector and physics processes, with particular emphasis given to the data expected from the first years of operation of the LHC at CERN

    Wnt signaling enhances neurogenesis and improves neurological function after focal ischemic injury.

    Get PDF
    Stroke potently stimulates cell proliferation in the subventricular zone of the lateral ventricles with subsequent neuroblast migration to the injured striatum and cortex. However, most of the cells do not survive and mature. Extracellular Wnt proteins promote adult neurogenesis in the neurogenic niches. The aim of the study was to examine the efficacy of Wnt signaling on neurogenesis and functional outcome after focal ischemic injury. Lentivirus expressing Wnt3a-HA (LV-Wnt3a-HA) or GFP (LV-GFP) was injected into the striatum or subventricular zone of mice. Five days later, focal ischemic injury was induced by injection of the vasoconstrictor endothelin-1 into the striatum of the same hemisphere. Treatment with LV-Wnt3a-HA into the striatum significantly enhanced functional recovery after ischemic injury and increased the number of BrdU-positive cells that differentiated into mature neurons in the ischemic striatum by day 28. Treatment with LV-Wnt3a-HA into the subventricular zone significantly enhanced functional recovery from the second day after injury and increased the number of immature neurons in the striatum and subventricular zone. This was accompanied by reduced dissemination of the neuronal injury. Our data indicate that Wnt signaling appears to contribute to functional recovery after ischemic injury by increasing neurogenesis or neuronal survival in the striatum

    DJ-1 based peptide, ND-13, promote functional recovery in mouse model of focal ischemic injury.

    No full text
    Stroke is a leading cause of death worldwide and inflicts serious long-term damage and disability. The vasoconstrictor Endothelin-1, presenting long-term neurological deficits associated with excitotoxicity and oxidative stress is being increasingly used to induce focal ischemic injury as a model of stroke. A DJ-1 based peptide named ND-13 was shown to protect against glutamate toxicity, neurotoxic insults and oxidative stress in various animal models. Here we focus on the benefits of treatment with ND-13 on the functional outcome of focal ischemic injury. Wild type C57BL/6 mice treated with ND-13, after ischemic induction in this model, showed significant improvement in motor function, including improved body balance and motor coordination, and decreased motor asymmetry. We found that DJ-1 knockout mice are more sensitive to Endothelin-1 ischemic insult than wild type mice, contributing thereby additional evidence to the widely reported relevance of DJ-1 in neuroprotection. Furthermore, treatment of DJ-1 knockout mice with ND-13, following Endothelin-1 induced ischemia, resulted in significant improvement in motor functions, suggesting that ND-13 provides compensation for DJ-1 deficits. These preliminary results demonstrate a possible basis for clinical application of the ND-13 peptide to enhance neuroprotection in stroke patients

    Neuroprotective Effect of a DJ-1 Based Peptide in a Toxin Induced Mouse Model of Multiple System Atrophy.

    No full text
    Multiple System Atrophy (MSA) is a sporadic neurodegenerative disorder characterized by parkinsonism, cerebellar ataxia and dysautonomia, in various combinations. In MSA with parkinsonism (MSA-P), the degeneration is mainly restricted to the substantia nigra pars compacta and putamen. Studies have identified alterations in DJ-1 (PARK7), a key component of the anti-oxidative stress response, in Parkinson's disease (PD) and MSA patients. Previously we have shown that a short DJ-1-based peptide named ND-13, protected cultured cells against neurotoxic insults and improved behavioral outcome in animal models of Parkinson's disease (PD). In this study, we used the 3-Nitropropionic acid (3-NP)-induced mouse model of MSA and treated the animals with ND-13 in order to evaluate its therapeutic effects. Our results show that ND-13 protects cultured cells against oxidative stress generated by the mitochondrial inhibitor, 3-NP. Moreover, we show that ND-13 attenuates nigrostriatal degeneration and improves performance in motor-related behavioral tasks in 3-NP-treated mice. Our findings suggest a rationale for using ND-13 as a promising therapeutic approach for treatment of MSA

    Changes in mitochondrial protein expression levels following ND-13 treatment.

    No full text
    <p>Ischemic wild type mice treated with ND-13 show significant change in protein expression levels compared to saline treated ischemic wild type mice. SDHAF4 protein levels increased after ND-13 treatment (A, FC = 107.4, p<0.001). Ubiquinone protein levels decreased after ND-13 treatment (B, FC = 3.5, p<0.01). Rhot2 protein levels decreased after ND-13 treatment (C, FC = 2.5, p<0.01). Usp35 protein levels decreased after ND-13 treatment (D, FC = 2.4, p<0.01). (Data is shown as mean ± SEM).</p

    DJ-1 KO mice show higher sensitivity and less spontaneous recovery after ischemic injury compared to C57BL/6 mice.

    No full text
    <p>DJ-1 KO mice show slower spontaneous recovery compared to C57BL/6 mice and are more sensitive to ischemic insult. DJ-1 KO mice spent significantly more time crossing the beam in the elevated bridge test (A, p<0.05) than wild type mice. DJ-1 KO mice show only 5% recovery from day 2 to days 7 after injury, compared to C57BL/6 mice that improved by 20% over the same period of time. Improvement was also noted in cylinder test 7 days after injury (B). (Data is shown as mean ± SEM).</p

    Neuroprotection in the ischemic striatum following LV-Wnt3a-HA injection into the SVZ 2 days after injury.

    No full text
    <p>A. DNA strand breaks are labeled by TUNEL staining and NeuN immunohistochemistry in the ischemic striatum. B. Wnt3a-HA significantly reduced the number of DNA fragmented cells in the striatum (<i>p</i><0.05). C. DCX<sup>+</sup> cells manifest extensive expression of BDNF in the ischemic striatum. D. Quantification of BDNF levels in the striatum using ELISA (<i>p</i><0.05).</p

    Effect of Wnt3a-HA treatment on neurogenesis 28 days after injury.

    No full text
    <p>A. A co-localized BrdU/NeuN cell is shown in the striatum. B–D. Wnt3a-HA injection into the striatum led to a significant increase in the number of newborn neurons in the striatum (<i>p</i><0.01). Treatment with Wnt3a-HA into the SVZ did not change the number of newborn neurons in the striatum. Number of newborn neurons (B), proliferating progenitors (C) and NeuN<sup>+</sup>/BrdU<sup>+</sup> (D) cells in the striatum and SVZ.</p
    corecore