Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Front Pharmacol ; 15: 1361415, 2024.
Article in English | MEDLINE | ID: mdl-38455960

ABSTRACT

Pharmacological education is crucial for healthcare professionals to safely manage medications and reduce errors. Traditional lecture-based learning (LBL) often struggles to address this complexity, whereas newer methods, such as flipped classrooms and problem-based learning, yield mixed results, particularly in pre-clinical contexts, owing to students' limited experience. Our nursing pharmacology course under LBL recorded a high failure rate of 37.8% and marginal passing scores across five cohorts (n = 849 students). An analysis using Bloom's taxonomy revealed significant gaps in higher-order cognitive skills. As a remedy, the course was transformed into a novel blended learning format that integrated question-based learning (QBL) to enhance critical thinking across all cognitive levels. This model blends asynchronous and synchronous learning, is tailored to individual needs in large classes, and fosters continuous, student-centric learning. The redesign markedly decreased the failure rate by approximately 2.8-fold and increased the average grade by 11.8 points among 426 students. It notably improved the pass rates in advanced cognitive categories, such as "Evaluate" and "Create" by 19.0% and 24.2%, respectively. Additionally, the blended course showed increased student engagement, reflecting a dynamic and effective learning environment that significantly elevated participation and academic outcomes at all cognitive levels. This study demonstrated the profound impact of blended learning in pharmacology. By integrating QBL with various teaching methods, it surpasses traditional lecture-based limitations, enhancing engagement and understanding of complex topics by nursing students. Notable improvements in foundational and advanced learning suggest its broader application in health professionals' education, effectively equipping students for clinical pharmacology challenges.

2.
Sci Adv ; 7(11)2021 03.
Article in English | MEDLINE | ID: mdl-33692101

ABSTRACT

No disease-modifying therapy is currently available for Parkinson's disease (PD), the second most common neurodegenerative disease. The long nonmotor prodromal phase of PD is a window of opportunity for early detection and intervention. However, we lack the pathophysiological understanding to develop selective biomarkers and interventions. By using a mutant α-synuclein selective-overexpression mouse model of prodromal PD, we identified a cell-autonomous selective Kv4 channelopathy in dorsal motor nucleus of the vagus (DMV) neurons. This functional remodeling of intact DMV neurons leads to impaired pacemaker function in vitro and in vivo, which, in turn, reduces gastrointestinal motility, a common early symptom of prodromal PD. We identify a chain of events from α-synuclein via a biophysical dysfunction of a specific neuronal population to a clinically relevant prodromal symptom. These findings will facilitate the rational design of clinical biomarkers to identify people at risk for developing PD.


Subject(s)
Channelopathies , Neurodegenerative Diseases , Parkinson Disease , Animals , Humans , Mice , Motor Neurons , Parkinson Disease/etiology , alpha-Synuclein/genetics
3.
Biochim Biophys Acta ; 1853(2): 422-30, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25450973

ABSTRACT

Mesenchymal stem cells are potent candidates in stroke therapy due to their ability to secrete protective anti-inflammatory cytokines and growth factors. We investigated the neuroprotective effects of human placental mesenchymal-like adherent stromal cells (PLX) using an established ischemic model of nerve growth factor (NGF)-differentiated pheochromocytoma PC12 cells exposed to oxygen and glucose deprivation (OGD) followed by reperfusion. Under optimal conditions, 2 × 105 PLX cells, added in a trans-well system, conferred 30-60% neuroprotection to PC12 cells subjected to ischemic insult. PC12 cell death, measured by LDH release, was reduced by PLX cells or by conditioned medium derived from PLX cells exposed to ischemia, suggesting the active release of factorial components. Since neuroprotection is a prominent function of the cytokine IL-6 and the angiogenic factor VEGF165, we measured their secretion using selective ELISA of the cells under ischemic or normoxic conditions. IL-6 and VEGF165 secretion by co-culture of PC12 and PLX cells was significantly higher under ischemic compared to normoxic conditions. Exogenous supplementation of 10 ng/ml each of IL-6 and VEGF165 to insulted PC12 cells conferred neuroprotection, reminiscent of the neuroprotective effect of PLX cells or their conditioned medium. Growth factors as well as co-culture conditioned medium effects were reduced by 70% and 20% upon pretreatment with 240 ng/ml Semaxanib (anti VEGF165) and/or 400 ng/ml neutralizing anti IL-6 antibody, respectively. Therefore, PLX-induced neuroprotection in ischemic PC12 cells may be partially explained by IL-6 and VEGF165 secretion. These findings may also account for the therapeutic effects seen in clinical trials after treatment with these cells.


Subject(s)
Cell Differentiation/drug effects , Interleukin-6/metabolism , Ischemia/pathology , Mesenchymal Stem Cells/cytology , Nerve Growth Factors/pharmacology , Neuroprotective Agents/metabolism , Placenta/cytology , Vascular Endothelial Growth Factor A/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Cell Adhesion/drug effects , Cell Count , Cell Death/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Cyclic N-Oxides/pharmacology , Female , Humans , Indoles/pharmacology , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Mice , PC12 Cells , Pregnancy , Pyrroles/pharmacology , Rats , Spin Labels
4.
J Neurotrauma ; 31(16): 1405-16, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24640955

ABSTRACT

Treatment of traumatic brain injury (TBI) is still an unmet need. Cell therapy by human umbilical cord blood (HUCB) has shown promising results in animal models of TBI and is under evaluation in clinical trials. HUCB contains different cell populations but to date, only mesenchymal stem cells have been evaluated for therapy of TBI. Here we present the neurotherapeutic effect, as evaluated by neurological score, using a single dose of HUCB-derived mononuclear cells (MNCs) upon intravenous (IV) administration one day post-trauma in a mouse model of closed head injury (CHI). Delayed (eight days post-trauma) intracerebroventricular administration of MNCs showed improved neurobehavioral deficits thereby extending the therapeutic window for treating TBI. Further, we demonstrated for the first time that HUCB-derived pan-hematopoietic CD45 positive (CD45(+)) cells, isolated by magnetic sorting and characterized by expression of CD45 and CD11b markers (96-99%), improved the neurobehavioral deficits upon IV administration, which persisted for 35 days. The therapeutic effect was in a direct correlation to a reduction in the lesion volume and decreased by pre-treatment of the cells with anti-human-CD45 antibody. At the site of brain injury, 1.5-2 h after transplantation, HUCB-derived cells were identified by near infrared scanning and immunohistochemistry using anti-human-CD45 and anti-human-nuclei antibodies. Nerve growth factor and vascular endothelial growth factor levels were differentially expressed in both ipsilateral and contralateral brain hemispheres, thirty-five days after CHI, measured by enzyme-linked immunosorbent assay. These findings indicate the neurotherapeutic potential of HUCB-derived CD45(+) cell population in a mouse model of TBI and propose their use in the clinical setting of human TBI.


Subject(s)
Brain Injuries/therapy , Cord Blood Stem Cell Transplantation/methods , Recovery of Function , Animals , Cell Separation , Disease Models, Animal , Flow Cytometry , Humans , Leukocyte Common Antigens/immunology , Male , Mice
5.
J Mol Neurosci ; 51(2): 249-61, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23233347

ABSTRACT

Umbilical cord blood (CB) stem cells have been proposed for cell-based therapeutic applications for diverse diseases of the CNS. We hypothesized that tissue-engineering strategies may extend the efficacy of these approaches by improving the long-term viability and function of stem cell-derived neuronal progenitors. To test our hypothesis, we explored the survival and differentiation of human CB-derived neuronal progenitors (HUCBNP) in a three-dimensional (3D) collagen construct. In contrast to two-dimensional culture conditions, the cells survived in 3D for an extended period of time of more than 2 months. Under 3D conditions, HUCBNP underwent spontaneous neuronal differentiation, which was further enhanced by treatment with neuronal conditioned medium (CM) and nerve growth factor (NGF). Neurite outgrowth, quantified by assessing the fractal dimension (D f) of the complex neuronal networks, was significantly enhanced under 3D conditions in the presence of CM/NGF, concomitant with a reduced expression of the early neuronal marker nestin (1.9-fold), and increased levels of mature neuronal markers such as MAP-2 (3.6-fold), ß-tubulin (1.5-fold), and neuronal specific enolase (6.6-fold) and the appearance of the synaptic marker synaptophysin. To assess the feasibility for clinical usage, HUCBNP were also isolated from frozen CB samples and cultured under 3D conditions. The data indicate the essential complete preservation of neurotrophic (survival) and neurotropic (neurite outgrowth) properties. In conclusion, 3D culture conditions are proposed as an essential step for both maintenance of CB neuronal progenitors in vitro and for investigating specific features of neuronal differentiation towards future use in regenerative therapy.


Subject(s)
Collagen/pharmacology , Fetal Blood/cytology , Mesenchymal Stem Cells/cytology , Neural Stem Cells/cytology , Neurogenesis , Tissue Scaffolds/chemistry , Cell Survival , Cryopreservation , Culture Media, Conditioned/pharmacology , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Growth Factor/pharmacology , Nestin/genetics , Nestin/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism , Tissue Engineering , Tubulin/genetics , Tubulin/metabolism
6.
PLoS One ; 7(11): e48803, 2012.
Article in English | MEDLINE | ID: mdl-23144978

ABSTRACT

Novel strategies that target the epidermal growth factor receptor (EGFR) have led to the clinical development of monoclonal antibodies, which treat metastatic colorectal cancer (mCRC) but only subgroups of patients with increased wild type KRAS and EGFR gene copy, respond to these agents. Furthermore, resistance to EGFR blockade inevitably occurred, making future therapy difficult. Novel bio-imaging (BOI) methods may assist in quantization of EGFR in mCRC tissue thus complementing the immunohistochemistry methodology, in guiding the future treatment of these patients. The aim of the present study was to explore the usefulness of near infrared-labeled EGF (EGF-NIR) for bio-imaging of CRC using in vitro and in vivo orthotopic tumor CRC models and ex vivo human CRC tissues. We describe the preparation and characterization of EGF-NIR and investigate binding, using BOI of a panel of CRC cell culture models resembling heterogeneity of human CRC tissues. EGF-NIR was specifically and selectively bound by EGFR expressing CRC cells, the intensity of EGF-NIR signal to background ratio (SBR) reflected EGFR levels, dose-response and time course imaging experiments provided optimal conditions for quantization of EGFR levels by BOI. EGF-NIR imaging of mice with HT-29 orthotopic CRC tumor indicated that EGF-NIR is more slowly cleared from the tumor and the highest SBR between tumor and normal adjacent tissue was achieved two days post-injection. Furthermore, images of dissected tissues demonstrated accumulation of EGF-NIR in the tumor and liver. EGF-NIR specifically and strongly labeled EGFR positive human CRC tissues while adjacent CRC tissue and EGFR negative tissues expressed weak NIR signals. This study emphasizes the use of EGF-NIR for preclinical studies. Combined with other methods, EGF-NIR could provide an additional bio-imaging specific tool in the standardization of measurements of EGFR expression in CRC tissues.


Subject(s)
Colorectal Neoplasms/pathology , Diagnostic Imaging/methods , Epidermal Growth Factor , ErbB Receptors/metabolism , Adult , Animals , Cell Line, Tumor , Humans , Mice , Molecular Probe Techniques , RNA Interference , Signal-To-Noise Ratio , Tumor Cells, Cultured
7.
Cell Signal ; 24(12): 2378-88, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22960610

ABSTRACT

The functions of nerve growth factor (NGF) in skeletal muscles physiology and pathology are not clear and call for an updated investigation. To achieve this goal we sought to investigate NGF-induced ERK1/2 phosphorylation and its role in the C2C12 skeletal muscle myoblasts and myotubes. RT-PCR and western blotting experiments demonstrated expression of p75(NTR), α9ß1 integrin, and its regulator ADAM12, but not trkA in the cells, as also found in gastrocnemius and quadriceps mice muscles. Both proNGF and ßNGF induced ERK1/2 phosphorylation, a process blocked by (a) the specific MEK inhibitor, PD98059; (b) VLO5, a MLD-disintegrin with relative selectivity towards α9ß1 integrin; and (c) p75(NTR) antagonists Thx-B and LM-24, but not the inactive control molecule backbone Thx. Upon treatment for 4 days with either anti-NGF antibody or VLO5 or Thx-B, the proliferation of myoblasts was decreased by 60-70%, 85-90% and 60-80% respectively, indicative of trophic effect of NGF which was autocrinically released by the cells. Exposure of myotubes to ischemic insult in the presence of ßNGF, added either 1h before oxygen-glucose-deprivation or concomitant with reoxygenation insults, resulted with about 20% and 33% myoprotection, an effect antagonized by VLO5 and Thx-B, further supporting the trophic role of NGF in C2C12 cells. Cumulatively, the present findings propose that proNGF and ßNGF-induced ERK1/2 phosphorylation in C2C12 cells by functional cooperation between p75(NTR) and α9ß1 integrin, which are involved in myoprotective effects of autocrine released NGF. Furthermore, the present study establishes an important trophic role of α9ß1 in NGF-induced signaling in skeletal muscle model, resembling the role of trkA in neurons. Future molecular characterization of the interactions between NGF receptors in the skeletal muscle will contribute to the understanding of NGF mechanism of action and may provide novel therapeutic targets.


Subject(s)
Integrins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Growth Factor/pharmacology , Receptors, Nerve Growth Factor/metabolism , ADAM Proteins/metabolism , ADAM12 Protein , Animals , Autocrine Communication , Cell Line , Cell Proliferation/drug effects , Flavonoids/pharmacology , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Phosphorylation/drug effects , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Nerve Growth Factor/genetics , Signal Transduction/drug effects
8.
Br Med Bull ; 104: 7-19, 2012.
Article in English | MEDLINE | ID: mdl-22988303

ABSTRACT

INTRODUCTION: Neural stem cells (NSCs) from specific brain areas or developed from progenitors of different sources are of therapeutic potential for neurodegenerative diseases. SOURCES OF DATA: Treatment strategies involve the (i) transplantation of exogenous NSCs; (ii) pharmacological modulations of endogenous NSCs and (iii) modulation of endogenous NSCs via the transplantation of exogenous NSCs. AREAS OF AGREEMENT: There is a consensus about the therapeutic potential of transplanted NSCs. The ability of NSCs to home into areas of central nervous system injury allows their delivery by intravenous injection. There is also a general agreement about the neuroprotective mechanisms of NSCs involving a 'bystander effect'. AREAS OF CONTROVERSY: Individual laboratories may be using phenotypically diverse NSCs, since these cells have been differentiated by a variety of neurotrophins and/or cultured on different ECM proteins, therefore differing in the expression of neuronal markers. GROWING POINTS: Optimization of the dose, delivery route, timing of administration of NSCs, their interactions with the immune system and combination therapies in conjunction with tissue engineered neural prostheses are under investigation. AREAS TIMELY FOR DEVELOPING RESEARCH: In-depth understanding of the biological properties of NSCs, including mechanisms of therapy, safety, efficacy and elimination from the organism. These areas are central for further use in cell therapy. CAUTIONARY NOTE: As long as critical safety and efficacy issues are not resolved, we need to be careful in translating NSC therapy from animal models to patients.


Subject(s)
Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Neurodegenerative Diseases/therapy , Neurogenesis , Brain/metabolism , Bystander Effect , Central Nervous System/metabolism , Central Nervous System Diseases/surgery , Central Nervous System Diseases/therapy , Humans , Neural Stem Cells/physiology , Stem Cell Transplantation/methods
9.
J Mol Neurosci ; 48(3): 526-40, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22678884

ABSTRACT

Pituitary adenylate cyclase activating peptide (PACAP), a potent neuropeptide which crosses the blood-brain barrier, is known to provide neuroprotection in rat stroke models of middle cerebral artery occlusion (MCAO) by mechanism(s) which deserve clarification. We confirmed that following i.v. injection of 30 ng/kg of PACAP38 in rats exposed to 2 h of MCAO focal cerebral ischemia and 48 h reoxygenation, 50 % neuroprotection was measured by reduced caspase-3 activity and volume of cerebral infarction. Similar neuroprotective effects were measured upon PACAP38 treatment of oxygen-glucose deprivation and reoxygenation of brain cortical neurons. The neuroprotection was temporally associated with increased expression of brain-derived neurotrophic factor, phosphorylation of its receptor-tropomyosin-related kinase receptor type B (trkB), activation of phosphoinositide 3-kinase and Akt, and reduction of extracellular signal-regulated kinases 1/2 phosphorylation. PACAP38 increased expression of neuronal markers beta-tubulin III, microtubule-associated protein-2, and growth-associated protein-43. PACAP38 induced stimulation of Rac and suppression of Rho GTPase activities. PACAP38 downregulated the nerve growth factor receptor (p75(NTR)) and associated Nogo-(Neurite outgrowth-A) receptor. Collectively, these in vitro and in vivo results propose that PACAP exhibits neuroprotective effects in cerebral ischemia by three mechanisms: a direct one, mediated by PACAP receptors, and two indirect, induced by neurotrophin release, activation of the trkB receptors and attenuation of neuronal growth inhibitory signaling molecules p75(NTR) and Nogo receptor.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/therapeutic use , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cell Hypoxia , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cerebral Cortex/cytology , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Glucose/pharmacology , Hypoxia-Ischemia, Brain/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Injections, Intravenous , Male , Nerve Tissue Proteins/genetics , Neurites/drug effects , Neurites/ultrastructure , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Phosphorylation/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/administration & dosage , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Protein Processing, Post-Translational/drug effects , Rats , Rats, Wistar , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/drug effects , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Reperfusion Injury/prevention & control , Signal Transduction/drug effects
10.
Int J Dev Neurosci ; 30(6): 465-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22677442

ABSTRACT

The goal of this study was to compare the neuroprotective properties of the L-type Ca²âº channel blockers, nimodipine and nifedipine, using nerve growth factor (NGF)-differentiated PC12 neuronal cultures exposed to oxygen-glucose deprivation (OGD) and trophic withdrawal-induced cell death. Nimodipine (1-100 µM) conferred 65±13% neuroprotection upon exposure to OGD and 35±6% neuroprotection towards different trophic withdrawal-induced cell death measured by lactate dehydrogenase and caspase 3 activities. The time window of nimodipine conferred neuroprotection was detected during the first 5h but not at longer OGD exposures. Nifedipine (1-100 µM), to a lower potency than nimodipine, conferred 30-55±8% neuroprotection towards OGD in PC12 cells and 29±5% in rat hypocampal slices, and 10±3% neuroprotection at 100 µM towards trophic withdrawal-induced PC12 cell death. The ability to demonstrate that nimodipine conferred neuroprotection in a narrow therapeutic time-window indicates that the OGD PC12 model mimics the in vivo models and therefore suitable for neuroprotective drug discovery and development.


Subject(s)
Cell Differentiation/drug effects , Glucose/deficiency , Nerve Growth Factor/pharmacology , Neuroprotective Agents/pharmacology , Nifedipine/pharmacology , Nimodipine/pharmacology , Analysis of Variance , Animals , Calcium/metabolism , Caspase 3/metabolism , Catecholamines/metabolism , Cell Hypoxia/drug effects , Hippocampus/drug effects , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Nerve Growth Factor/metabolism , PC12 Cells/drug effects , Rats
11.
Toxins (Basel) ; 3(11): 1420-32, 2011 11.
Article in English | MEDLINE | ID: mdl-22174978

ABSTRACT

In Israel, Vipera xantina palestinae (V.x.p.) is the most common venomous snake, accounting for several hundred cases of envenomation in humans and domestic animals every year, with a mortality rate of 0.5 to 2%. In this review we will briefly address the research developments relevant to our present understanding of the structure and function of V.x.p. venom with emphasis on venom disintegrins. Venom proteomics indicated the presence of four families of pharmacologically active compounds: (i) neurotoxins; (ii) hemorrhagins; (iii) angioneurin growth factors; and (iv) different types of integrin inhibitors. Viperistatin, a α1ß1selective KTS disintegrin and VP12, a α2ß1 selective C-type lectin were discovered. These snake venom proteins represent promising tools for research and development of novel collagen receptor selective drugs. These discoveries are also relevant for future improvement of antivenom therapy towards V.x.p. envenomation.


Subject(s)
Viper Venoms/chemistry , Animals , Antivenins/therapeutic use , Humans , Integrins/antagonists & inhibitors , Nerve Growth Factor/analysis , Neurotoxins/analysis , Proteome , Vascular Endothelial Growth Factor A/analysis , Viper Venoms/analysis , Viperidae
12.
Arch Ital Biol ; 149(2): 233-45, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21701995

ABSTRACT

Stem cells have an extremely high potential to treat many devastating diseases, including neuronal injuries. Albeit the need for human neuronal stem cells, their quantities are very limited by relying on early human embryos as the main source. Therefore, progenitors of other origins, such as human umbilical cord blood (CB) are being considered. In the last decade, various populations isolated from the CB were reported to differentiate in vitro towards a neural phenotype. The conditions to induce the cell differentiation are not conclusive and may include addition of chemicals, cytokines and growth factors, including the nerve growth factor (NGF). Some CB cells were found to express the TrkANGF receptor, suggesting an endogenous role for this growth factor also in the CB environment. The ability of CB and derived stem cell populations to protect against neurological deficits was shown, both in vitro and in vivo, in models of ischemic brain injuries. In rodent models of stroke, heatstroke, brain trauma and brain damage at birth, CB cells either by intravenous injection or intrastriatal transplantation, were found to reduce the infarct size and the neurological deficits caused by the injury. The restorative effects of CB were suggested to be mediated by mechanisms other than cell replacement. Some of the proposed mechanisms involve reduced inflammation, nerve fiber reorganization by trophic actions, increased cell survival and enhanced angiogenesis. Furthermore, treatment with CB was found to have a therapeutic window of days compared with the present 36 hour window for the treatment of stroke with clinically available tools such as recombinant tissue plasminogen activator. Considering the encouraging results with whole CB and derived cells transplantation in ischemic injury models and since CB is widely available and have been used clinically, they may be an excellent source of cells for treatment of human brain ischemic disorders.


Subject(s)
Brain Injuries/prevention & control , Brain Ischemia/surgery , Cord Blood Stem Cell Transplantation/methods , Fetal Blood/cytology , Hematopoietic Stem Cells/physiology , Animals , Brain Injuries/etiology , Brain Ischemia/complications , Cell Differentiation , Humans , Nerve Growth Factor/metabolism , Nerve Growth Factor/therapeutic use , Neuroprotective Agents/metabolism , Neuroprotective Agents/therapeutic use
13.
Int J Mol Sci ; 11(9): 3513-28, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20957109

ABSTRACT

The use of stem cells for reparative medicine was first proposed more than three decades ago. Hematopoietic stem cells from bone marrow, peripheral blood and human umbilical cord blood (CB) have gained major use for treatment of hematological indications. CB, however, is also a source of cells capable of differentiating into various non-hematopoietic cell types, including neural cells. Several animal model reports have shown that CB cells may be used for treatment of neurological injuries. This review summarizes the information available on the origin of CB-derived neuronal cells and the mechanisms proposed to explain their action. The potential use of stem/progenitor cells for treatment of ischemic brain injuries is discussed. Issues that remain to be resolved at the present stage of preclinical trials are addressed.


Subject(s)
Brain Ischemia/surgery , Cord Blood Stem Cell Transplantation , Animals , Humans , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Neurogenesis
14.
Best Pract Res Clin Haematol ; 23(2): 291-303, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20837341

ABSTRACT

Regenerative medicine is the process of creating functional tissue with the aid of stem cells, to repair loss of organ function. Possible targets for regenerative medicine include orthopaedic, cardiac, hepatic, pancreatic and central nervous system (CNS) applications. Umbilical cord blood (CB) has established itself as a legitimate source for haematopoietic stem cell transplantation. It is also considered an accessible and less immunogenic source for mesenchymal, unrestricted somatic and for other stem cells with pluri/multipotent properties. The latter are capable of differentiating into a wide variety of cell types including bone, cartilage, cardiomyocytes and neural. They also possess protective abilities that may contribute to tissue repair even if in vitro differentiation is excluded. In view of the absence of treatment for many devastating diseases, the elucidation of non-haematopoietic applications for CB will facilitate the development of pioneering relevant cell therapy approaches. This review focusses on current studies using human CB-derived cells for regenerative medicine.


Subject(s)
Cell Differentiation , Fetal Blood , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Regeneration , Cell- and Tissue-Based Therapy/methods , Cord Blood Stem Cell Transplantation/methods , Humans , Organ Specificity , Regenerative Medicine/methods
15.
Vascul Pharmacol ; 53(3-4): 107-14, 2010.
Article in English | MEDLINE | ID: mdl-20434587

ABSTRACT

Cumulative evidences suggest that nerve growth factor (NGF) promotes angiogenic effects such as proliferation and migration of endothelial cells (ECs) from different vascular beds, induces capillary sprouting in chorioallantoic membrane and improves in vivo vascularization in a hind-limb ischemic model. In the present study, we sought to investigate the signaling properties of NGF in a microcapillary ECs model compared to those of a neuronal model. NGF-induced phosphorylation of signaling molecules Erk1/2, Akt and PLCgamma were measured using Western blotting and compared between mouse NGF (mNGF) and snake venom NGF analogues. NGFs-induced signaling was TrkA mediated as evident by inhibition with the TrkA antagonist K252a. NGF and its analogues-induced signaling in ECs were characterized by a transient effect in contrast to a prolonged stimulation in neuronal cells. The potency of mouse, cobra and viper NGFs to induce Erk1/2 phosphorylation in ECs was higher than in neurons. In ECs, mNGF exhibited the highest efficacy of stimulation of Erk1/2 phosphorylation, followed by viper and cobra NGFs. The efficacy of stimulation of Erk1/2 phosphorylation measured with neurons was opposite from that in ECs. NGF-induced temporal signaling differences between ECs and neurons may explain the dual vascular and neurotrophic effects of this growth factor.


Subject(s)
Brain/blood supply , Endothelial Cells/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Growth Factor/metabolism , Phospholipase C gamma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Carbazoles/pharmacology , Cells, Cultured , Chorioallantoic Membrane/metabolism , Elapid Venoms/pharmacology , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Activation , Indole Alkaloids/pharmacology , Mice , Neurons/metabolism , Phosphorylation , Rats , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/metabolism , Signal Transduction , Viper Venoms/pharmacology
16.
Mol Cell Biochem ; 339(1-2): 201-13, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20148355

ABSTRACT

Nerve growth factor (NGF) supports the survival and differentiation of sympathetic and sensory neurons and is also mitogenic for a variety of tumors. K252a, an antagonist of NGF receptor TrkA, was previously used as a pharmacological tool to study NGF actions and as a lead compound for developing anti-tumor drugs. Since recently, NGF was characterized as an angiogenic factor, we sought to investigate the angiostatic properties of K252a on endothelial cells (ECs). For this purpose, we used a murine brain microcapillary ECs model in which we found autocrine release of NGF in the culture medium and activation of TrkA receptor-induced downstream signaling molecules Erk1/2, Akt, and PLCgamma. In this model, we demonstrated the angiostatic property of K252a based on its ability to affect several important angiogenic steps. K252a, but not its cell membrane impermeable analogue K252b at 100 nM: (i) inhibited the proliferation of the ECs by 45 +/- 9%; (ii) reduced by 70 +/- 4% the migration of the ECs measured in a wound-closure model; (iii) reduced by 29 +/- 9% the formation of tube-like structures of the ECs cultured on Matrigel; (iv) stimulated by 100 +/- 25% the collagen deposition by the ECs, a process responsible for the increased endothelial barrier functions expressed by 22 +/- 5% reduction of paracellular permeability and by 17 +/- 3% elevation of transendothelial electrical resistance. These data suggest that NGF/TrkA may represent a target for the development of novel, K252a-derived multikinase inhibitors drugs with anti-tumor and angiostatic dual activities.


Subject(s)
Brain/drug effects , Brain/metabolism , Carbazoles/pharmacology , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Indole Alkaloids/pharmacology , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/metabolism , Animals , Blotting, Western , Brain/cytology , Cell Differentiation/drug effects , Cell Membrane Permeability/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Collagen/metabolism , Drug Combinations , Endothelial Cells/metabolism , Laminin/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Phosphorylation/drug effects , Proteoglycans/metabolism , RNA, Messenger/genetics , Receptor, trkA/genetics , Reverse Transcriptase Polymerase Chain Reaction
17.
In Vitro Cell Dev Biol Anim ; 46(5): 469-76, 2010 May.
Article in English | MEDLINE | ID: mdl-20112076

ABSTRACT

Endothelial cells (ECs) from different vascular beds not only display common characteristics but are also quite heterogeneous in terms of expression and secretion of neuro-angiogenic factors, which may help explain some of their distinct physiological roles. We investigated by RT-PCR the gene expression, by PC12 bioassay the neurotropic activity, and by ELISAs the levels of NGF and FGF-2 using conditioned medium collected from cultures of ECs derived from myocardial and cerebral capillaries. While NGF was expressed and released by both cell types, FGF-2 was expressed and released solely by the brain but not heart ECs. Oxygen-glucose deprivation (ischemic) insult blocked NGF secretion from heart and brain ECs and inhibited by 70% the secretion of FGF-2 from brain ECs. We propose that the differential expression of NGF and FGF-2 in heart and brain EC cultures reflect heterogeneity on demand of the microcapillary components and the surrounding microenvironment for a proper tissue-specific homeostasis.


Subject(s)
Endothelium, Vascular/physiology , Fibroblast Growth Factor 2/biosynthesis , Nerve Growth Factor/biosynthesis , Animals , Biological Assay , Brain/metabolism , Brain/physiology , Cell Line , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression/physiology , Heart/physiology , Hypoxia/metabolism , Hypoxia/physiopathology , Mice , Myocardium/metabolism , Nerve Growth Factor/physiology , PC12 Cells/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction
18.
J Mol Neurosci ; 41(1): 183-92, 2010 May.
Article in English | MEDLINE | ID: mdl-20012228

ABSTRACT

Nerve growth factor (NGF) was recently characterized as an angiogenic factor inducing proliferation, migration, and capillary sprouting in endothelial cells (ECs) of different vascular beds. While NGF neuroprotective effects on neurons were described, its survival-inducing effects on brain capillary ECs were not yet addressed. Using a model of oxygen-glucose deprivation (OGD) followed by reoxygenation, we demonstrated that NGF conferred protection in brain capillary ECs. These cells express TrkA and p75(NTR) receptors and respond to NGF by stimulation of Erk1/2 phosphorylation and stimulation of proliferation and migration. The NGF protective effect was dose-dependent, inhibited by NGF/TrkA antagonist, K252a, and required presence of NGF during both OGD and reoxygenation phases while the major protective effect was related to decreased cell death during the reoxygenation phase. A causal relationship was found between NGF-induced protection and attenuation of OGD-induced Erk1/2 phosphorylation, supporting the death-promoting role of insult-induced Erk1/2 phosphorylation in the brain capillary ECs. These results emphasize the importance of NGF in the process of EC survival in response to ischemic injury and suggest fine-tuning regulation of Erk1/2 phosphorylation, extending the neuroprotective impact of NGF from sympathetic neuroendocrine cells to brain capillary ECs as the other element in the neurovascular tandem.


Subject(s)
Brain Ischemia/metabolism , Endothelial Cells , Glucose/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Growth Factor/pharmacology , Oxygen/metabolism , Animals , Cell Death/physiology , Cell Movement/physiology , Cell Proliferation , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression , Mice , Neuroprotective Agents/pharmacology , Phosphorylation/drug effects , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Signal Transduction/physiology
19.
J Mol Neurosci ; 37(3): 225-37, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18629654

ABSTRACT

There is a paucity of quantitative methods for evaluating the morphological differentiation of neuronal cells in a three-dimensional (3-D) system to assist in quality control of neural tissue engineering constructs for use in reparative medicine. Neuronal cells tend to aggregate in the 3-D scaffolds, hindering the application of two-dimensional (2-D) morphological methods to quantitate neuronal differentiation. To address this problem, we developed a stable transfectant green fluorescence protein (GFP)-PC12 neuronal cell model, in which the differentiation process in 3-D can be monitored with high sensitivity by fluorescence microscopy. Under 2-D conditions, the green cells showed collagen adherence, round morphology, proliferation properties, expression of the nerve growth factor (NGF) receptors TrkA and p75(NTR), stimulation of extracellular signal-regulated kinase phosphorylation by NGF and were able to differentiate in a dose-dependent manner upon NGF treatment, like wild-type (wt)-PC12 cells. When grown within 3-D collagen gels, upon NGF treatment, the GFP-PC12 cells differentiated, expressing long neurite outgrowths. We describe here a new validated method to measure NGF-induced differentiation in 3-D. Having properties similar to those of wt-PC12 and an ability to grow and differentiate in 3-D structures, these highly visualized GFP-expressing PC12 cells may serve as an ideal model for investigating various aspects of differentiation to serve in neural engineering.


Subject(s)
Cell Differentiation/physiology , Collagen/metabolism , Gels/metabolism , Green Fluorescent Proteins/metabolism , Neurons/physiology , PC12 Cells , Animals , Cell Proliferation , Green Fluorescent Proteins/genetics , Neurons/cytology , PC12 Cells/cytology , PC12 Cells/physiology , Rats , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism
20.
Exp Neurol ; 216(1): 83-94, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19070617

ABSTRACT

Human umbilical cord blood (HUCB) is a valuable source for cell therapy since it confers neuroprotection in stroke animal models. However, the responsible sub-populations remain to be established and the mechanisms involved are unknown. To explore HUCB neuroprotective properties in a PC12 cell-based ischemic neuronal model, we used an HUCB mononuclear-enriched population of collagen-adherent cells, which can be differentiated in vitro into a neuronal phenotype (HUCBNP). Upon co-culture with insulted-PC12 cells, HUCBNP conferred approximately 30% neuroprotection, as evaluated by decreased lactate dehydrogenase and caspase-3 activities. HUCBNP decreased by 95% the level of free radicals in the insulted-PC12 cells, in correlation with the appearance of antioxidants, as measured by changes in the oxidation-reduction potential of the medium using cyclic-voltammetry. An increased level of nerve growth factor (NGF), vascular endothelial growth factor and basic fibroblast growth factor in the co-culture medium was temporally correlated with a -medium neuroprotection effect, which was partially abolished by heat denaturation. HUCBNP-induced neuroprotection was correlated with changes in gene expression of these neurotrophic factors, while blocked by K252a, an antagonist of the TrkA/NGF receptor. These findings indicate that HUCBNP-induced neuroprotection involves antioxidant(s) and neurotrophic factors, which, by paracrine and/or autocrine interactions between the insulted-PC12 and the HUCBNP cells, conferred neuroprotection.


Subject(s)
Angiogenic Proteins/metabolism , Antioxidants/metabolism , Cytoprotection/physiology , Fetal Blood/cytology , Nerve Growth Factors/metabolism , Neurons/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Free Radicals/metabolism , Humans , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Hypoxia-Ischemia, Brain/therapy , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Oxidation-Reduction , Oxidative Stress/physiology , PC12 Cells , Phenotype , Rats , Stem Cells/cytology , Stroke/metabolism , Stroke/physiopathology , Stroke/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...