Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cells ; 12(12)2023 06 13.
Article in English | MEDLINE | ID: mdl-37371088

ABSTRACT

The enormous, 2-3-million-year evolutionary expansion of hominin neocortices to the current enormity enabled humans to take over the planet. However, there appears to have been a glitch, and it occurred without a compensatory expansion of the entorhinal cortical (EC) gateway to the hippocampal memory-encoding system needed to manage the processing of the increasing volume of neocortical data converging on it. The resulting age-dependent connectopathic glitch was unnoticed by the early short-lived populations. It has now surfaced as Alzheimer's disease (AD) in today's long-lived populations. With advancing age, processing of the converging neocortical data by the neurons of the relatively small lateral entorhinal cortex (LEC) inflicts persistent strain and high energy costs on these cells. This may result in their hyper-release of harmless Aß1-42 monomers into the interstitial fluid, where they seed the formation of toxic amyloid-ß oligomers (AßOs) that initiate AD. At the core of connectopathic AD are the postsynaptic cellular prion protein (PrPC). Electrostatic binding of the negatively charged AßOs to the positively charged N-terminus of PrPC induces hyperphosphorylation of tau that destroys synapses. The spread of these accumulating AßOs from ground zero is supported by Aß's own production mediated by target cells' Ca2+-sensing receptors (CaSRs). These data suggest that an early administration of a strongly positively charged, AßOs-interacting peptide or protein, plus an inhibitor of CaSR, might be an effective AD-arresting therapeutic combination.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Neurons/metabolism , Entorhinal Cortex/metabolism , Prion Proteins/metabolism
2.
Pharm Res ; 39(7): 1497-1507, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35704250

ABSTRACT

PURPOSE: We have recently demonstrated the brain-delivery of an Amyloid-ß oligomer (Aßo)-binding peptide-therapeutic fused to the BBB-crossing single domain antibody FC5. The bi-functional fusion protein, FC5-mFc-ABP (KG207-M) lowered both CSF and brain Aß levels after systemic dosing in transgenic mouse and rat models of Alzheimer's disease (AD). For development as a human therapeutic, we have humanized and further engineered the fusion protein named KG207-H. The purpose of the present study was to carry out comparative PK/PD studies of KG207-H in wild type rat and beagle dogs (middle-aged and older) to determine comparability of systemic PK and CSF exposure between rodent species and larger animals with more complex brain structure such as dogs. METHOD: Beagle dogs were used in this study as they accumulate cerebral Aß with age, as seen in human AD patients, and can serve as a model of sporadic AD. KG207-H (5 to 50 mg/kg) was administered intravenously and serum and CSF samples were serially collected for PK studies and to assess target engagement. KG207-H and Aß levels were quantified using multiplexed selected reaction monitoring mass spectrometry. RESULTS: After systemic dosing, KG207-H demonstrated similar serum pharmacokinetics in rats and dogs. KG207-H appeared in the CSF in a time- and dose-dependent manner with similar kinetics, indicating CNS exposure. Further analyses revealed a dose-dependent inverse relationship between CSF KG207-H and Aß levels in both species indicating target engagement. CONCLUSION: This study demonstrates translational attributes of BBB-crossing Aß-targeting biotherapeutic KG207-H in eliciting a pharmacodynamic response, from rodents to larger animal species.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Dogs , Mice , Mice, Transgenic , Rats
3.
J Cereb Blood Flow Metab ; 42(5): 788-801, 2022 05.
Article in English | MEDLINE | ID: mdl-34378436

ABSTRACT

In vivo biomarker abnormalities provide measures to monitor therapeutic interventions targeting amyloid-ß pathology as well as its effects on downstream processes associated with Alzheimer's disease pathophysiology. Here, we applied an in vivo longitudinal study design combined with imaging and cerebrospinal fluid biomarkers, mirroring those used in human clinical trials to assess the efficacy of a novel brain-penetrating anti-amyloid fusion protein treatment in the McGill-R-Thy1-APP transgenic rat model. The bi-functional fusion protein consisted of a blood-brain barrier crossing single domain antibody (FC5) fused to an amyloid-ß oligomer-binding peptide (ABP) via Fc fragment of mouse IgG (FC5-mFc2a-ABP). A five-week treatment with FC5-mFc2a-ABP (loading dose of 30 mg/Kg/iv followed by 15 mg/Kg/week/iv for four weeks) substantially reduced brain amyloid-ß levels as measured by positron emission tomography and increased the cerebrospinal fluid amyloid-ß42/40 ratio. In addition, the 5-week treatment rectified the cerebrospinal fluid neurofilament light chain concentrations, resting-state functional connectivity, and hippocampal atrophy measured using magnetic resonance imaging. Finally, FC5-mFc2a-ABP (referred to as KG207-M) treatment did not induce amyloid-related imaging abnormalities such as microhemorrhage. Together, this study demonstrates the translational values of the designed preclinical studies for the assessment of novel therapies based on the clinical biomarkers providing tangible metrics for designing early-stage clinical trials.


Subject(s)
Alzheimer Disease , Amyloidosis , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Biomarkers , Longitudinal Studies , Mice , Positron-Emission Tomography , Rats
4.
PLoS One ; 16(5): e0250568, 2021.
Article in English | MEDLINE | ID: mdl-33970919

ABSTRACT

The development of effective therapies as well as early, molecular diagnosis of Alzheimer's disease is impeded by the lack of understanding of the underlying pathological mechanisms. Metabolomics studies of body fluids as well as brain tissues have shown major changes in metabolic profiles of Alzheimer's patients. However, with analysis performed at the late stages of the disease it is not possible to distinguish causes and consequence. The mouse model APP/PS1 expresses a mutant amyloid precursor protein resulting in early Amyloid ß (Aß) accumulation as well as many resulting physiological changes including changes in metabolic profile and metabolism. Analysis of metabolic profile of cerebrospinal fluid (CSF) and blood of APP/PS1 mouse model can provide information about metabolic changes in these body fluids caused by Aß accumulation. Using our novel method for analysis of correlation and mathematical ranking of significant correlations between metabolites in CSF and blood, we have explored changes in metabolite correlation and connectedness in APP/PS1 and wild type mice. Metabolites concentration and correlation changes in CSF, blood and across the blood brain barrier determined in this work are affected by the production of amyloid plaque. Metabolite changes observed in the APP/PS1 mouse model are the response to the mutation causing plaque formation, not the cause for the plaque suggesting that they are less relevant in the context of early treatment and prevention then the metabolic changes observed only in humans.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Cerebrospinal Fluid/metabolism , Magnetic Resonance Spectroscopy/methods , Metabolome , Presenilin-1/genetics , Serum/metabolism , Alzheimer Disease/blood , Alzheimer Disease/cerebrospinal fluid , Animals , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
5.
Biochem Cell Biol ; 96(4): 391-406, 2018 08.
Article in English | MEDLINE | ID: mdl-29370536

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of morbidity and mortality worldwide. Due to its high incidence rate and often long-term sequelae, TBI contributes significantly to increasing costs of health care expenditures annually. Unfortunately, advances in the field have been stifled by patient and injury heterogeneity that pose a major challenge in TBI prevention, diagnosis, and treatment. In this review, we briefly discuss the causes of TBI, followed by its prevalence, classification, and pathophysiology. The current imaging detection methods and animal models used to study brain injury are examined. We discuss the potential use of molecular markers in detecting and monitoring the progression of TBI, with particular emphasis on microRNAs as a novel class of molecular modulators of injury and its repair in the neural tissue.


Subject(s)
Biomarkers/analysis , Brain Injuries, Traumatic , Functional Neuroimaging , MicroRNAs/therapeutic use , Animals , Brain/diagnostic imaging , Brain Injuries, Traumatic/diagnosis , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/therapy , Disease Models, Animal , Humans
6.
FASEB J ; 30(5): 1927-40, 2016 05.
Article in English | MEDLINE | ID: mdl-26839377

ABSTRACT

Receptor mediated transcytosis harnessing the cellular uptake and transport of natural ligands across the blood-brain barrier (BBB) has been identified as a means for antibody delivery to the CNS. In this study, we characterized bispecific antibodies in which a BBB-crossing antibody fragment FC5 was used as a BBB carrier. Cargo antibodies were either a high-affinity, selective antibody antagonist of the metabotropic glutamate receptor-1 (BBB-mGluR1), a widely abundant CNS target, or an IgG that does not bind the CNS target (BBB-NiP). Both BBB-NiP and BBB-mGluR1 demonstrated a similar 20-fold enhanced rate of transcytosis across an in vitro BBB model compared with mGluR1 IgG fused to a control antibody fragment. All 3 bispecific antibodies exhibited identical pharmacokinetics in vivo Comparative assessment of BBB-NiP and BBB-mGluR1 revealed that, whereas their serum pharmacokinetics and BBB penetration were identical, their central disposition (brain levels) and elimination (cerebrospinal fluid levels) were widely different, due to central target-mediated removal of the mGluR1-engaging antibody. Central mGluR1 target engagement after systemic administration was demonstrated by a dose-dependent inhibition of mGluR-1-mediated thermal hyperalgesia and by colocalization of the antibody with thalamic neurons involved in mGluR1-mediated pain processing. We demonstrate the feasibility of targeting central G-protein-coupled receptors using a BBB-crossing bispecific antibody approach and emerging principles that govern brain distribution and disposition of these antibodies. These data will be important for designing safe and selective CNS antibody therapeutics.-Webster, C. I., Caram-Salas, N., Haqqani, A. S., Thom, G., Brown, L., Rennie, K., Yogi, A., Costain, W., Brunette, E., Stanimirovic, D. B. Brain penetration, target engagement, and disposition of the blood-brain barrier-crossing bispecific antibody antagonist of metabotropic glutamate receptor type 1.


Subject(s)
Antibodies, Bispecific/pharmacology , Brain/metabolism , Pain/drug therapy , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Analgesics , Animals , Biological Products/metabolism , Biological Transport , Blood-Brain Barrier/metabolism , Brain/drug effects , Camelidae , Cell Membrane , HEK293 Cells , Hot Temperature/adverse effects , Humans , Immunoconjugates/metabolism , Immunoglobulin G/immunology , Pain/etiology , Protein Engineering/methods , Rats , Receptors, Metabotropic Glutamate/metabolism
7.
Stem Cell Rev Rep ; 10(2): 251-68, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24415130

ABSTRACT

Brain injury continues to be one of the leading causes of disability worldwide. Despite decades of research, there is currently no pharmacologically effective treatment for preventing neuronal loss and repairing the brain. As a result, novel therapeutic approaches, such as cell-based therapies, are being actively pursued to repair tissue damage and restore neurological function after injury. In this study, we examined the neuroprotective potential of amniotic fluid (AF) single cell clones, engineered to secrete glial cell derived neurotrophic factor (AF-GDNF), both in vitro and in a surgically induced model of brain injury. Our results show that pre-treatment with GDNF significantly increases cell survival in cultures of AF cells or cortical neurons exposed to hydrogen peroxide. Since improving the efficacy of cell transplantation depends on enhanced graft cell survival, we investigated whether AF-GDNF cells seeded on polyglycolic acid (PGA) scaffolds could enhance graft survival following implantation into the lesion cavity. Encouragingly, the AF-GDNF cells survived longer than control AF cells in serum-free conditions and continued to secrete GDNF both in vitro and following implantation into the injured motor cortex. AF-GDNF implantation in the acute period following injury was sufficient to activate the MAPK/ERK signaling pathway in host neural cells in the peri-lesion area, potentially boosting endogenous neuroprotective pathways. These results were complemented with promising trends in beam walk tasks in AF-GDNF/PGA animals during the 7 day timeframe. Further investigation is required to determine whether significant behavioural improvement can be achieved at a longer timeframe.


Subject(s)
Amniotic Fluid/cytology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Stem Cell Transplantation , Stem Cells/physiology , Animals , Brain Injuries/pathology , Brain Injuries/physiopathology , Brain Injuries/therapy , Cell Survival , Cells, Cultured , Female , Gene Expression , Humans , Hydrogen Peroxide/pharmacology , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Motor Cortex/pathology , Neural Stem Cells/physiology , Oxidants/pharmacology , Oxidative Stress , Prostheses and Implants , Psychomotor Performance , Tissue Scaffolds
8.
Biochem Cell Biol ; 91(5): 271-86, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24032676

ABSTRACT

There is a need for improved therapy for acquired brain injury, which has proven resistant to treatment by numerous drugs in clinical trials and continues to represent one of the leading causes of disability worldwide. Research into cell-based therapies for the treatment of brain injury is growing rapidly, but the ideal cell source has yet to be determined. Subpopulations of cells found in amniotic fluid, which is readily obtained during routine amniocentesis, can be easily expanded in culture, have multipotent differentiation capacity, are non-tumourigenic, and avoid the ethical complications associated with embryonic stem cells, making them a promising cell source for therapeutic purposes. Beneficial effects of amniotic fluid cell transplantation have been reported in various models of nervous system injury. However, evidence that amniotic fluid cells can differentiate into mature, functional neurons in vivo and incorporate into the existing circuitry to replace lost or damaged neurons is lacking. The mechanisms by which amniotic fluid cells improve outcomes after experimental nervous system injury remain unclear. However, studies reporting the expression and release of neurotrophic, angiogenic, and immunomodulatory factors by amniotic fluid cells suggest they may provide neuroprotection and (or) stimulate endogenous repair and remodelling processes in the injured nervous system. In this paper, we address recent research related to the neuronal differentiation of amniotic fluid-derived cells, the therapeutic efficacy of these cells in animal models of nervous system injury, and the possible mechanisms mediating the positive outcomes achieved by amniotic fluid cell transplantation.


Subject(s)
Amniotic Fluid/cytology , Brain Injuries/therapy , Cell- and Tissue-Based Therapy/methods , Multipotent Stem Cells/transplantation , Amniocentesis , Animals , Cell Differentiation , Humans , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Neurons/cytology , Stroke/therapy , Tissue Engineering/methods
9.
Stem Cells Int ; 2012: 721538, 2012.
Article in English | MEDLINE | ID: mdl-23093978

ABSTRACT

The amniotic membrane (AM) and amniotic fluid (AF) have a long history of use in surgical and prenatal diagnostic applications, respectively. In addition, the discovery of cell populations in AM and AF which are widely accessible, nontumorigenic and capable of differentiating into a variety of cell types has stimulated a flurry of research aimed at characterizing the cells and evaluating their potential utility in regenerative medicine. While a major focus of research has been the use of amniotic membrane and fluid in tissue engineering and cell replacement, AM- and AF-derived cells may also have capabilities in protecting and stimulating the repair of injured tissues via paracrine actions, and acting as vectors for biodelivery of exogenous factors to treat injury and diseases. Much progress has been made since the discovery of AM and AF cells with stem cell characteristics nearly a decade ago, but there remain a number of problematic issues stemming from the inherent heterogeneity of these cells as well as inconsistencies in isolation and culturing methods which must be addressed to advance the field towards the development of cell-based therapies. Here, we provide an overview of the recent progress and future perspectives in the use of AM- and AF-derived cells for therapeutic applications.

10.
Stem Cells Int ; 2012: 607161, 2012.
Article in English | MEDLINE | ID: mdl-22792116

ABSTRACT

The usage of stem cells is a promising strategy for the repair of damaged tissue in the injured brain. Recently, amniotic fluid (AF) cells have received a lot of attention as an alternative source of stem cells for cell-based therapies. However, the success of this approach relies significantly on proper interactions between graft and host tissue. In particular, the reestablishment of functional brain networks requires formation of gap junctions, as a key step to provide sufficient intercellular communication. In this study, we show that AF cells express high levels of CX43 (GJA1) and are able to establish functional gap junctions with cortical cultures. Furthermore, we report an induction of Cx43 expression in astrocytes following injury to the mouse motor cortex and demonstrate for the first time CX43 expression at the interface between implanted AF cells and host brain cells. These findings suggest that CX43-mediated intercellular communication between AF cells and cortical astrocytes may contribute to the reconstruction of damaged tissue by mediating modulatory, homeostatic, and protective factors in the injured brain and hence warrants further investigation.

11.
Brain Res ; 1373: 79-90, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21138733

ABSTRACT

Previous work in our laboratory indicated that cholinergic denervation by intraventricular infusion of 192-IgG-saporin on postnatal day 7 (N192S) reduced the number of cells in the dentate gyrus expressing doublecortin, a marker for immature neuroblasts. In addition, there was a suggestion that N192S impaired the neurogenic response to environmental enrichment (EE). The purpose of the present study was to further characterize the impact of N192S on the proliferation, differentiation and survival of newborn cells in the dentate gyrus. After 42 days in EE or standard housing, all rats received injections of 5-bromo-2-deoxyuridine (BrdU) to label dividing cells. They were sacrificed either one day (to assess cell proliferation) or 28 days later (to assess survival and differentiation of BrdU-labelled cells). EE failed to increase neurogenesis, thereby preventing determination of the effects of N192S on EE-induced neurogenesis. However, N192S by itself reduced the number of BrdU(+) cells 1 day after BrdU exposure, but did not alter the number of cells expressing the cell cycle marker Ki-67. The number of BrdU(+) cells 28 days after BrdU exposure was not affected by N192S. Confocal analysis of BrdU(+) cells double-immunofluorescently stained to detect NeuN or S100B indicated that N192S did not alter the proportion of new cells that adopted a neuronal or glial identity. The most plausible explanation for these results is that N192S accelerates the death of newborn cells, but does not change their overall survival rate or phenotypic differentiation.


Subject(s)
Antibodies, Monoclonal/toxicity , Cholinergic Agents/toxicity , Hippocampus/physiopathology , Neurogenesis/physiology , Prosencephalon/drug effects , Ribosome Inactivating Proteins, Type 1/toxicity , Acetylcholinesterase/metabolism , Age Factors , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , Doublecortin Domain Proteins , Doublecortin Protein , Female , Ki-67 Antigen/metabolism , Male , Microtubule-Associated Proteins/metabolism , Nerve Growth Factors/metabolism , Neurogenesis/drug effects , Neuropeptides/metabolism , Prosencephalon/growth & development , Prosencephalon/injuries , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor/metabolism , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Saporins , Statistics as Topic
12.
J Pineal Res ; 47(4): 313-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19796045

ABSTRACT

It was previously shown that pinealectomy causes delayed loss of pyramidal neurons in rat hippocampal layers CA1/3 and that this is reversed by melatonin supplementation. Here, we used immunohistologic detection of doublecortin, a protein expressed in newborn neurons, to determine if melatonin supplementation promotes neurogenesis after pinealectomy. It was found that melatonin supplementation significantly increased the number of doublecortin immunoreactive neurons in the dentate gyrus over the postsurgical intervals of 2, 4, 6, 8, 10 and 17 months. The increase was most evident at 6 months postsurgery and thereafter, and was apparent despite a severe decline in doublecortin-labeled cells over the 17 month postsurgical interval in all groups of rats. Doublecortin immunoreactive cells were not observed in the pyramidal layer itself. These results indicate that melatonin promotes neurogenesis in the dentate gyrus of pinealectomized rats. However, it is equivocal that these newborn neurons migrate to the pyramidal layer and account for the reappearance of neurons at this location in these rats. This study provides further evidence for a role of melatonin in promoting neurogenesis, adding another role to its already remarkably pleiotropic profile. The scope and significance of this newly discovered role remains to be determined.


Subject(s)
Central Nervous System Depressants/pharmacology , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Melatonin/pharmacology , Neurogenesis/drug effects , Pineal Gland/surgery , Animals , Doublecortin Domain Proteins , Doublecortin Protein , Immunohistochemistry , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Rats , Rats, Sprague-Dawley
13.
Brain Res ; 1252: 172-82, 2009 Feb 03.
Article in English | MEDLINE | ID: mdl-19084506

ABSTRACT

Intraventricular injections of 192 IgG saporin in 7-day-old rat severely reduced hippocampal cholinergic innervation as reflected by both decreased acetylcholinesterase staining and immunoreactivity for the p75 neurotrophin receptor. It was determined if this altered the effects of environmental enrichment on spatial learning, hippocampal CA1 cell cytoarchitecture as reflected by the Golgi stain, and neurogenesis in the dentate gyrus as indicated by doublecortin immunoreactivity. At weaning, lesioned and control rats were either group housed in large, environmentally enriched cages or housed two per standard cage for 42 days. When subsequently assessed with a working-memory spatial navigation task, both lesioned and control rats showed enhanced learning as a result of enrichment. Quantitative analysis of Golgi stained sections indicated that enrichment did not affect CA1 dendritic branching, total dendritic length or dendritic spine density. However, the lesion reduced the number of apical branches, spine density on intermediate to distal apical dendrites, and the length of basal branches. It also reduced the number of doublecortin immunoreactive neurons in the dentate gyrus and appeared to prevent their increase due to environmental enrichment. It is concluded that developmental cholinergic lesioning does not attenuate neurobehavioral plasticity, at least as reflected by the behavioral consequences of enrichment. It does, however, attenuate neurogenesis in the dentate gyrus, like adult-inflicted cholinergic lesions. As previously found for cortical neurons, it also reduces CA1 pyramidal cell dendritic complexity and spine density in adulthood. The results have implications for the loss of synapses that occurs in both developmental and aging-related brain disorders involving cholinergic dysfunction.


Subject(s)
Acetylcholine/metabolism , Brain Injuries/physiopathology , Environment , Hippocampus/cytology , Neurogenesis , Prosencephalon/injuries , Prosencephalon/physiopathology , Analysis of Variance , Animals , Antibodies, Monoclonal/pharmacology , Behavior, Animal/physiology , Brain Injuries/chemically induced , Brain Injuries/therapy , Dendritic Spines , Doublecortin Domain Proteins , Doublecortin Protein , Female , Hippocampus/physiopathology , Immunohistochemistry , Male , Maze Learning , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Neurotoxins/pharmacology , Prosencephalon/anatomy & histology , Pyramidal Cells/physiopathology , Pyramidal Cells/ultrastructure , Rats , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins
14.
J Pineal Res ; 44(2): 149-56, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18289166

ABSTRACT

Melatonin attenuates the short-term consequences of brain ischemia in several animal models. However, there is scant information regarding its efficacy for improving the long-term outcome. To further address that issue, we subjected gerbils to 5-min bilateral carotid occlusion. Some gerbils received acute peri-surgical administration of melatonin while others received continuous melatonin in their water. The gerbils' brains were histologically assessed at 20 wk postsurgery. Chronic but not acute melatonin attenuated ischemia-induced hyperactivity at 3 days postsurgery. Twenty weeks postsurgery, the ischemic gerbils showed varying degrees of bilateral loss of hippocampal CA1 pyramidal cells and elevation of glial fibrillary acidic protein immunoreactivity there. Both the cell loss and the immunoreactivity were markedly asymmetrical for some gerbils. Neither acute nor chronic melatonin altered this pattern of CA1 cell loss and glial immunoreactivity increase. Ischemia increased the number of CA1 cells that were immunoreactive for doublecortin (DCX), a marker for newborn neurons. This increase in CA1 DCX expression was not affected by either melatonin treatment. However, both acute and chronic melatonin reduced the number of DCX immunoreactive neurons in the dentate gyrus. Thus, neither acute nor chronic melatonin altered the long-term neural outcome of forebrain ischemia, although chronic administration seemed to attenuate the short-term behavioral effect. It is suggested that persistently high brain levels of melatonin may be essential for long-term neuroprotection against ischemia. The possibility that melatonin may modulate hippocampal neurogenesis merits further exploration both in normal animals and in models of brain insult.


Subject(s)
Behavior, Animal/physiology , Brain Ischemia/metabolism , Cell Differentiation/drug effects , Melatonin/administration & dosage , Neurons/drug effects , Prosencephalon/metabolism , Animals , Behavior, Animal/drug effects , Brain Ischemia/pathology , Cell Count , Cell Differentiation/physiology , Disease Models, Animal , Drug Administration Schedule , Gerbillinae , Male , Melatonin/blood , Melatonin/physiology , Neurons/pathology , Prosencephalon/drug effects , Prosencephalon/pathology , Random Allocation , Stroke/metabolism , Time Factors , Treatment Outcome
15.
J R Soc Interface ; 4(16): 767-76, 2007 Oct 22.
Article in English | MEDLINE | ID: mdl-17626002

ABSTRACT

The interaction between sheep and the nematode Teladorsagia circumcincta is one of the best understood of all host-parasite interactions. Following infection, there is considerable variation among lambs in the number of nematode eggs produced, the number of early fourth-stage larvae and the number of adult worms in the mucosa. These traits have a high variance to mean ratio (i.e. they are overdispersed or aggregated among hosts), they are skewed and approximately negative binomially distributed. The sources of overdispersion are differences among lambs in the ingestion of infective larvae and the immune response. Both forces can produce aggregation but their relative importance is unknown. The key components of variation can be identified by variance analysis. The sum of the average effects of polymorphic genes is known as additive genetic variation and this increases essentially from zero at one month of age to quite high values at six months of age. The major mechanism underlying genetic variation appears to be the differences among individuals in immune responses. Two of the major sources of variation in immune responses are differences in antigen recognition and differences in the type of cytokines produced. Genes that influence both these sources of variation are associated with differences in resistance to nematode infection. Therefore, much of the heterogeneity among animals in parasite transmission appears to be due to genetic variation in immune responsiveness.


Subject(s)
Gastrointestinal Diseases/veterinary , Genetic Predisposition to Disease/genetics , Genetic Variation , Nematode Infections/veterinary , Sheep Diseases/genetics , Sheep Diseases/parasitology , Animals , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/parasitology , Nematode Infections/genetics , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...