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








Publication year range
1.
Proc Natl Acad Sci U S A ; 121(29): e2401420121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38995966

ABSTRACT

Cerebral (Aß) plaque and (pTau) tangle deposition are hallmarks of Alzheimer's disease (AD), yet are insufficient to confer complete AD-like neurodegeneration experimentally. Factors acting upstream of Aß/pTau in AD remain unknown, but their identification could enable earlier diagnosis and more effective treatments. T cell abnormalities are emerging AD hallmarks, and CD8 T cells were recently found to mediate neurodegeneration downstream of tangle deposition in hereditary neurodegeneration models. The precise impact of T cells downstream of Aß/pTau, however, appears to vary depending on the animal model. Our prior work suggested that antigen-specific memory CD8 T ("hiT") cells act upstream of Aß/pTau after brain injury. Here, we examine whether hiT cells influence sporadic AD-like pathophysiology upstream of Aß/pTau. Examining neuropathology, gene expression, and behavior in our hiT mouse model we show that CD8 T cells induce plaque and tangle-like deposition, modulate AD-related genes, and ultimately result in progressive neurodegeneration with both gross and fine features of sporadic human AD. T cells required Perforin to initiate this pathophysiology, and IFNγ for most gene expression changes and progression to more widespread neurodegenerative disease. Analogous antigen-specific memory CD8 T cells were significantly elevated in the brains of human AD patients, and their loss from blood corresponded to sporadic AD and related cognitive decline better than plasma pTau-217, a promising AD biomarker candidate. We identify an age-related factor acting upstream of Aß/pTau to initiate AD-like pathophysiology, the mechanisms promoting its pathogenicity, and its relevance to human sporadic AD.


Subject(s)
Alzheimer Disease , CD8-Positive T-Lymphocytes , Disease Models, Animal , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Mice , Humans , Plaque, Amyloid/pathology , Plaque, Amyloid/immunology , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Brain/pathology , Brain/immunology , Male , Interferon-gamma/metabolism , Interferon-gamma/immunology , Aging/immunology , Immunologic Memory , Memory T Cells/immunology , Perforin/metabolism , Perforin/genetics , Female
2.
bioRxiv ; 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38328072

ABSTRACT

Cerebral (Aß) plaque and (pTau) tangle deposition are hallmarks of Alzheimer's disease (AD), yet are insufficient to confer complete AD-like neurodegeneration experimentally. Factors acting upstream of Aß/pTau in AD remain unknown, but their identification could enable earlier diagnosis and more effective treatments. T cell abnormalities are emerging AD hallmarks, and CD8 T cells were recently found to mediate neurodegeneration downstream of tangle deposition in hereditary neurodegeneration models. The precise impact of T cells downstream of Aß/fibrillar pTau, however, appears to vary depending on the animal model used. Our prior work suggested that antigen-specific memory CD8 T (" hi T") cells act upstream of Aß/pTau after brain injury. Here we examine whether hi T cells influence sporadic AD-like pathophysiology upstream of Aß/pTau. Examining neuropathology, gene expression, and behavior in our hi T mouse model we show that CD8 T cells induce plaque and tangle-like deposition, modulate AD-related genes, and ultimately result in progressive neurodegeneration with both gross and fine features of sporadic human AD. T cells required Perforin to initiate this pathophysiology, and IFNγ for most gene expression changes and progression to more widespread neurodegenerative disease. Analogous antigen-specific memory CD8 T cells were significantly elevated in the brains of human AD patients, and their loss from blood corresponded to sporadic AD and related cognitive decline better than plasma pTau-217, a promising AD biomarker candidate. Our work is the first to identify an age-related factor acting upstream of Aß/pTau to initiate AD-like pathophysiology, the mechanisms promoting its pathogenicity, and its relevance to human sporadic AD. Significance Statement: This study changes our view of Alzheimer's Disease (AD) initiation and progression. Mutations promoting cerebral beta-amyloid (Aß) deposition guarantee rare genetic forms of AD. Thus, the prevailing hypothesis has been that Aß is central to initiation and progression of all AD, despite contrary animal and patient evidence. We show that age-related T cells generate neurodegeneration with compelling features of AD in mice, with distinct T cell functions required for pathological initiation and neurodegenerative progression. Knowledge from these mice was applied to successfully predict previously unknown features of human AD and generate novel tools for its clinical management.

3.
Front Neurol ; 11: 557269, 2020.
Article in English | MEDLINE | ID: mdl-33424735

ABSTRACT

The incidence of autism spectrum disorders (ASD) and attention deficit hyperactivity disorder (ADHD), which frequently co-occur, are both rising. The causes of ASD and ADHD remain elusive, even as both appear to involve perturbation of the gut-brain-immune axis. CD103 is an integrin and E-cadherin receptor most prominently expressed on CD8 T cells that reside in gut, brain, and other tissues. CD103 deficiency is well-known to impair gut immunity and resident T cell function, but it's impact on neurodevelopmental disorders has not been examined. We show here that CD8 T cells influence neural progenitor cell function, and that CD103 modulates this impact both directly and potentially by controlling CD8 levels in brain. CD103 knockout (CD103KO) mice exhibited a variety of behavioral abnormalities, including superior cognitive performance coupled with repetitive behavior, aversion to novelty and social impairment in females, with hyperactivity with delayed learning in males. Brain protein markers in female and male CD103KOs coincided with known aspects of ASD and ADHD in humans, respectively. Surprisingly, CD103 deficiency also decreased age-related cognitive decline in both sexes, albeit by distinct means. Together, our findings reveal a novel role for CD103 in brain developmental function, and identify it as a unique factor linking ASD and ADHD etiology. Our data also introduce a new animal model of combined ASD and ADHD with associated cognitive benefits, and reveal potential therapeutic targets for these disorders and age-related cognitive decline.

4.
J Neurol Neurophysiol ; 5(3)2014 May.
Article in English | MEDLINE | ID: mdl-25346865

ABSTRACT

T-lymphocytes have been previously implicated in protecting dopaminergic neurons in the substantianigra from induced cell death. However, the role of T-cells in neurodegenerative models such as Parkinson's disease (PD) has not been fully elucidated. To examine the role of T-lymphocytes on motor behavior in the 6-hydroxydopamine (6-OHDA) unilateral striatal partial lesion PD rat model, we assessed progression of hemi-parkinsonian lesions in the substantia nigra, induced by 6-OHDA striatal injections, in athymic rats (RNU-/-, T-lymphocyte-deficient) as compared to RNU-/+ rats (phenotypically normal). Motor skills were determined by the cylinder and D-amphetamine sulfate-induced rotational behavioral tests. Cylinder behavioral test showed no significant difference between unilaterally lesioned RNU-/- and RNU-/+ rats. However both unilaterally lesioned RNU-/- and RNU-/+ rats favored the use of the limb ipsilateral to lesion. Additionally, amphetamine-induced rotational test revealed greater rotational asymmetry in RNU-/- rats compared to RNU-/+ rats at two- and six-week post-lesion. Quantitative immunohistochemistry confirmed loss of striatal TH-immunopositive fibers in RNU-/- and RNU-/+ rat, as well as blood-brain-barrier changes associated with PD that may influence passage of immune cells into the central nervous system in RNU-/- brains. Specifically, GFAP immunopositive cells were decreased, as were astrocytic end-feet (AQP4) contacting blood vessels (laminin) in the lesioned relative to contralateral striatum. Flow cytometric analysis in 6-OHDA lesioned RNU-/+rats revealed increased CD4+ and decreased CD8+ T cells specifically within lesioned brain. These results suggest that both major T cell subpopulations are significantly and reciprocally altered following 6-OHDA-lesioning, and that global T cell deficiency exacerbates motor behavioral defects in this rat model of PD.

5.
Cancer Immunol Immunother ; 63(9): 911-24, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24893855

ABSTRACT

BACKGROUND: Cancer vaccines reproducibly cure laboratory animals and reveal encouraging trends in brain tumor (glioma) patients. Identifying parameters governing beneficial vaccine-induced responses may lead to the improvement of glioma immunotherapies. CD103(+) CD8 T cells dominate post-vaccine responses in human glioma patients for unknown reasons, but may be related to recent thymic emigrant (RTE) status. Importantly, CD8 RTE metrics correlated with beneficial immune responses in vaccinated glioma patients. METHODS: We show by flow cytometry that murine and human CD103(+) CD8 T cells respond better than their CD103(-) counterparts to tumor peptide-MHC I (pMHC I) stimulation in vitro and to tumor antigens on gliomas in vivo. RESULTS: Glioma responsive T cells from mice and humans both exhibited intrinsic de-sialylation-affecting CD8 beta. Modulation of CD8 T cell sialic acid with neuraminidase and ST3Gal-II revealed de-sialylation was necessary and sufficient for promiscuous binding to and stimulation by tumor pMHC I. Moreover, de-sialylated status was required for adoptive CD8 T cells and lymphocytes to decrease GL26 glioma invasiveness and increase host survival in vivo. Finally, increased tumor ST3Gal-II expression correlated with clinical vaccine failure in a meta-analysis of high-grade glioma patients. CONCLUSIONS: Taken together, these findings suggest that de-sialylation of CD8 is required for hyper-responsiveness and beneficial anti-glioma activity by CD8 T cells. Because CD8 de-sialylation can be induced with exogenous enzymes (and appears particularly scarce on human T cells), it represents a promising target for clinical glioma vaccine improvement.


Subject(s)
Antigens, CD/immunology , Brain Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/pharmacology , Dendritic Cells/immunology , Glioma/therapy , Integrin alpha Chains/immunology , Animals , Antigens, CD/metabolism , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Female , Glioblastoma/immunology , Glioblastoma/metabolism , Glioblastoma/therapy , Glioma/immunology , Glioma/metabolism , Humans , Immunotherapy, Adoptive/methods , Integrin alpha Chains/metabolism , Mice , Mice, Inbred C57BL , Neuraminidase/metabolism , Neuraminidase/pharmacology , Sialyltransferases/metabolism , Sialyltransferases/pharmacology , beta-Galactoside alpha-2,3-Sialyltransferase
6.
J Neurosurg ; 117(4): 705-11, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22920963

ABSTRACT

OBJECT: The prognosis of patients with glioblastoma who present with multifocal disease is not well documented. The objective of this study was to determine whether multifocal disease on initial presentation is associated with worse survival. METHODS: The authors retrospectively reviewed records of 368 patients with newly diagnosed glioblastoma and identified 47 patients with multifocal tumors. Each patient with a multifocal tumor was then matched with a patient with a solitary glioblastoma on the basis of age, Karnofsky Performance Scale (KPS) score, and extent of resection, using a propensity score matching methodology. Radiation and temozolomide treatments were also well matched between the 2 cohorts. Kaplan-Meier estimates and log-rank tests were used to compare patient survival. RESULTS: The incidence of multifocal tumors was 12.8% (47/368). The median age of patients with multifocal tumors was 61 years, 76.6% had KPS scores ≥ 70, and 87.2% underwent either a biopsy or partial resection of their tumors. The 47 patients with multifocal tumors were almost perfectly matched on the basis of age (p = 0.97), extent of resection (p = 1.0), and KPS score (p = 0.80) compared with 47 patients with a solitary glioblastoma. Age (>65 years), partial resection or biopsy, and low KPS score (<70) were associated with worse median survival within the multifocal group. In the multifocal group, 19 patients experienced tumor progression on postradiation therapy MRI, compared with 11 patients (26.8%) with tumor progression in the unifocal group (p = 0.08). Patients with multifocal tumors experienced a significantly shorter median overall survival of 6 months (95% CI 4-10 months), compared with the 11-month median survival (95% CI 10-19 months) of the matched solitary glioblastoma group (p = 0.02, log-rank test). Two-year survival rates were 4.3% for patients with multifocal tumors and 29.0% for the unifocal cohort. Patients with newly diagnosed multifocal tumors were found to have an almost 2-fold increase in the hazard of death compared with patients with solitary glioblastoma (hazard ratio 1.8, 95% CI 1.1-3.1; p = 0.02). Tumor samples were analyzed for expression of phosphorylated mitogen-activated protein kinase, phosphatase and tensin homolog, O(6)-methylguanine-DNA methyltransferase, laminin ß1 and ß2, as well as epidermal growth factor receptor amplification, and no significant differences in expression profile between the multifocal and solitary glioblastoma groups was found. CONCLUSIONS: Patients with newly diagnosed multifocal glioblastoma on presentation experience significantly worse survival than patients with solitary glioblastoma. Patients with multifocal tumors continue to pose a therapeutic challenge in the temozolomide era and magnify the challenges faced while treating patients with malignant gliomas.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Glioblastoma/diagnosis , Glioblastoma/therapy , Neoplasms, Multiple Primary/diagnosis , Neoplasms, Multiple Primary/therapy , Aged , Antineoplastic Agents/therapeutic use , Brain Neoplasms/mortality , Case-Control Studies , Cohort Studies , Combined Modality Therapy , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Glioblastoma/mortality , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasms, Multiple Primary/mortality , Neurosurgical Procedures , Prognosis , Radiotherapy , Retrospective Studies , Survival Rate , Temozolomide , Treatment Outcome
7.
PLoS One ; 5(6): e10974, 2010 Jun 07.
Article in English | MEDLINE | ID: mdl-20539758

ABSTRACT

BACKGROUND: Small populations of highly tumorigenic stem-like cells (cancer stem cells; CSCs) can exist within, and uniquely regenerate cancers including malignant brain tumors (gliomas). Many aspects of glioma CSCs (GSCs), however, have been characterized in non-physiological settings. METHODS: We found gene expression similarity superiorly defined glioma "stemness", and revealed that GSC similarity increased with lower tumor grade. Using this method, we examined stemness in human grade IV gliomas (GBM) before and after dendritic cell (DC) vaccine therapy. This was followed by gene expression, phenotypic and functional analysis of murine GL26 tumors recovered from nude, wild-type, or DC-vaccinated host brains. RESULTS: GSC similarity was specifically increased in post-vaccine GBMs, and correlated best to vaccine-altered gene expression and endogenous anti-tumor T cell activity. GL26 analysis confirmed immune alterations, specific acquisition of stem cell markers, specifically enhanced sensitivity to anti-stem drug (cyclopamine), and enhanced tumorigenicity in wild-type hosts, in tumors in proportion to anti-tumor T cell activity. Nevertheless, vaccine-exposed GL26 cells were no more tumorigenic than parental GL26 in T cell-deficient hosts, though they otherwise appeared similar to GSCs enriched by chemotherapy. Finally, vaccine-exposed GBM and GL26 exhibited relatively homogeneous expression of genes expressed in progenitor cells and/or differentiation. CONCLUSIONS: T cell activity represents an inducible physiological process capable of proportionally enriching GSCs in human and mouse gliomas. Stem-like gliomas enriched by strong T cell activity, however, may differ from other GSCs in that their stem-like properties may be disassociated from increased tumor malignancy and heterogeneity under specific host immune conditions.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Neoplastic Stem Cells/cytology , T-Lymphocytes, Cytotoxic/cytology , Animals , Base Sequence , Brain Neoplasms/genetics , DNA Primers , Flow Cytometry , Fluorescent Antibody Technique , Glioma/genetics , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
8.
Brain Res ; 1230: 290-302, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18674521

ABSTRACT

The blood-brain tumor barrier (BTB) significantly limits delivery of therapeutic concentrations of chemotherapy to brain tumors. A novel approach to selectively increase drug delivery is pharmacologic modulation of signaling molecules that regulate BTB permeability, such as those in cGMP signaling. Here we show that oral administration of sildenafil (Viagra) and vardenafil (Levitra), inhibitors of cGMP-specific PDE5, selectively increased tumor capillary permeability in 9L gliosarcoma-bearing rats with no significant increase in normal brain capillaries. Tumor-bearing rats treated with the chemotherapy agent, adriamycin, in combination with vardenafil survived significantly longer than rats treated with adriamycin alone. The selective increase in tumor capillary permeability appears to be mediated by a selective increase in tumor cGMP levels and increased vesicular transport through tumor capillaries, and could be attenuated by iberiotoxin, a selective inhibitor for calcium-dependent potassium (K(Ca)) channels, that are effectors in cGMP signaling. The effect by sildenafil could be further increased by simultaneously using another BTB "opener", bradykinin. Collectively, this data demonstrates that oral administration of PDE5 inhibitors selectively increases BTB permeability and enhances anti-tumor efficacy for a chemotherapeutic agent. These findings have significant implications for improving delivery of anti-tumor agents to brain tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Glioma/drug therapy , Glioma/metabolism , Phosphodiesterase 5 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Animals , Autoradiography , Blood Pressure/drug effects , Brain Chemistry/drug effects , Brain Neoplasms/pathology , Capillaries/pathology , Capillary Permeability/drug effects , Cyclic GMP/blood , Cyclic GMP/metabolism , Female , Glioma/pathology , Imidazoles/pharmacology , Microscopy, Electron, Transmission , Neovascularization, Pathologic/pathology , Piperazines/pharmacology , Purines/pharmacology , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Sildenafil Citrate , Sucrose/metabolism , Sulfones/pharmacology , Survival Analysis , Tight Junctions/drug effects , Tight Junctions/ultrastructure , Triazines/pharmacology , Vardenafil Dihydrochloride
9.
Clin Cancer Res ; 14(12): 4002-9, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559623

ABSTRACT

PURPOSE: The blood-brain tumor barrier (BTB) significantly limits the delivery of chemotherapeutics to brain tumors. Nitric oxide (NO) is involved in the regulation of cerebral vascular permeability. We investigated the effects of NO donors, L-arginine and hydroxyurea, on BTB permeability in 9L gliosarcoma-bearing Fischer rats. EXPERIMENTAL DESIGN: The rats implanted with 9L gliosarcoma were dosed orally with hydroxyurea and L-arginine. BTB permeability, defined by the unidirectional transport constant, Ki, for [14C]sucrose was measured. The expression of neural and endothelial NO synthase (NOS) in tumors and normal brain tissue was examined. Further, the levels of NO, L-citrulline, and cGMP in the tumor and normal brain tissue were measured. RESULTS: Oral administration of l-arginine or hydroxyurea significantly increased BTB permeability when compared with the nontreated control. The selective effects were abolished by iberiotoxin, an antagonist of calcium-dependent potassium (KCa) channel that is a cGMP pathway effector. The expression of endothelial NOS, but not neural NOS, was higher in tumor vessels than in those of normal brain. Moreover, the levels of NO, L-citrulline, a byproduct of NO formation from L-arginine, and cGMP were enhanced in the tumor tissue by oral administration of L-arginine and/or hydroxyurea. CONCLUSIONS: Oral administration of L-arginine or hydroxyurea selectively increased tumor permeability, which is likely mediated by alteration in cGMP levels. The findings suggest that use of oral NO donors may be a strategy to enhance the delivery of chemotherapeutics to malignant brain tumors.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Neoplasms/pathology , Capillary Permeability/drug effects , Glioma/pathology , Nitric Oxide Donors/pharmacology , Administration, Oral , Animals , Arginine/administration & dosage , Arginine/pharmacology , Blood-Brain Barrier/physiology , Brain Neoplasms/metabolism , Brain Neoplasms/physiopathology , Capillary Permeability/physiology , Citrulline/metabolism , Cyclic GMP/metabolism , Drug Delivery Systems , Drug Evaluation, Preclinical , Drug Synergism , Female , Glioma/metabolism , Glioma/physiopathology , Hydroxyurea/administration & dosage , Hydroxyurea/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
10.
Neurobiol Dis ; 30(3): 343-352, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18417351

ABSTRACT

The degeneration of neurons in the mammalian brain is commonly associated with the division of cells located in the damaged area. The aim of the present study has been to characterise the phenotype of newly born cells in the striatum of adult rats following 6-hydroxydopamine lesion of the nigro-striatal pathway. Newborn cells were identified through labelling with either bromodeoxyuridine or retrovirus encoding green fluorescence protein. We report here that the overwhelming majority of these cells have glial characteristics. In order to promote the generation of new neurons we retrovirally introduced either the noggin or neurogenin2 genes into newborn cells following the 6-hydroxydopamine lesion. Transduction with neurogenin2 resulted in the production of cells resembling neuroblasts, however these cells did not appear to survive. Noggin transduction did not result in the generation of new neurons, but interestingly, greatly increased the number of oligodendrocytes generated from newborn cells.


Subject(s)
Cell Proliferation , Corpus Striatum/physiology , Gene Transfer Techniques , Oxidopamine/toxicity , Substantia Nigra/physiology , Animals , Cell Proliferation/drug effects , Corpus Striatum/cytology , Corpus Striatum/drug effects , Female , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Substantia Nigra/cytology , Substantia Nigra/drug effects
11.
Exp Cell Res ; 313(6): 1172-80, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17291494

ABSTRACT

Parkinson's Disease (PD) is a debilitating motor function disorder due primarily to a loss of midbrain dopaminergic neurons and a subsequent reduction in dopaminergic innervation of the striatum. Several attempts have been made to generate dopaminergic neurons from progenitor cell populations in vitro for potential use in cell replacement therapy for PD. However, expanding cells from fetal brain with retained potential for dopaminergic differentiation has proven to be difficult. In this study, we sought to generate mesencephalic dopaminergic (mesDA) neurons from an expanded population of fetal mouse ventral midbrain (VM) progenitors through the use of retroviral gene delivery. We over-expressed Ngn2 and Nurr1, two genes present in the ventral midbrain and important for normal development of mesDA neurons, in multi-passaged neurosphere-expanded midbrain progenitors. We show that over-expression of Ngn2 in these progenitors results in increased neuronal differentiation but does not promote mesDA formation. We also show that over-expression of Nurr1 alone is sufficient to generate tyrosine hydroxylase (TH) expressing cells with an immature morphology, however the cells do not express any additional markers of mesDA neurons. Over-expression of Nurr1 and Ngn2 in combination generates morphologically mature TH-expressing neurons that also express additional mesencephalic markers.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , DNA-Binding Proteins/physiology , Dopamine/metabolism , Mesencephalon/cytology , Nerve Tissue Proteins/physiology , Neurons/physiology , Stem Cells/physiology , Transcription Factors/physiology , Animals , Cell Differentiation , Mesencephalon/metabolism , Mice , Neurons/enzymology , Nuclear Receptor Subfamily 4, Group A, Member 2 , Stem Cells/enzymology , Tyrosine 3-Monooxygenase/metabolism
12.
J Neurosci Res ; 75(3): 330-43, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14743446

ABSTRACT

The Notch-DSL signaling system, consisting of multiple receptors and ligands, inhibits neurogenesis and promotes gliogenesis during embryonic development, but the specific function of the various ligands and receptors at later developmental stages are unknown. Here, we examined the expression pattern of four Delta, Serrate and Lag-2 (DSL) ligands, Jagged1, Jagged2, Delta-like1 (Dl1) and Delta-like 3 (Dl3), in late embryonic and postnatal rat brain by in situ hybridization. In late embryos, Jagged1, Dl1 and Dl3 mRNAs were present in the periventricular germinal epithelia, but this expression diminished during postnatal ages. Jagged1 mRNA was also expressed in the inner aspect of the dentate gyrus at early postnatal times. Dl3 was detectable in the external granule cell layer (EGL) of the cerebellum, another site of postnatal neurogenesis. Jagged2 mRNA was expressed in virtually all postnatal neurons. Jagged1 mRNA was highly expressed in several brain nuclei during postnatal development, with lower levels of expression in other grey matter regions. In white matter, Dl1 and Dl3 mRNAs were expressed during the first week of postnatal development but only the expression of Dl1 mRNA persisted through the second week. Dl1 mRNA was present at lower levels throughout grey matter during the first few weeks of development. Jagged1 mRNA was expressed in blood vessels, choroid plexus, and menninges throughout development and in the adult. Jagged2 mRNA was transiently expressed in cerebral blood vessels and choroid plexus during the first postnatal week. Taken together, these results support multiple and differing roles for the various ligands during and after central nervous system (CNS) development.


Subject(s)
Brain/growth & development , Carrier Proteins/biosynthesis , Membrane Proteins/biosynthesis , Protein Biosynthesis , Proteins , Animals , Animals, Newborn , Brain/blood supply , Brain/embryology , Calcium-Binding Proteins , Cell Differentiation , Embryo, Mammalian , Embryonic and Fetal Development , Female , Gene Expression Regulation, Developmental , In Situ Hybridization , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Jagged-1 Protein , Jagged-2 Protein , Male , Neurons/metabolism , RNA, Messenger , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Serrate-Jagged Proteins , Stem Cells/metabolism
13.
Dev Biol ; 264(2): 309-22, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14651920

ABSTRACT

The identification of the genes regulating neural progenitor cell (NPC) functions is of great importance to developmental neuroscience and neural repair. Previously, we combined genetic subtraction and microarray analysis to identify genes enriched in neural progenitor cultures. Here, we apply a strategy to further stratify the neural progenitor genes. In situ hybridization demonstrates expression in the central nervous system germinal zones of 54 clones so identified, making them highly relevant for study in brain and neural progenitor development. Using microarray analysis we find 73 genes enriched in three neural stem cell (NSC)-containing populations generated under different conditions. We use the custom microarray to identify 38 "stemness" genes, with enriched expression in the three NSC conditions and present in both embryonic stem cells and hematopoietic stem cells. However, comparison of expression profiles from these stem cell populations indicates that while there is shared gene expression, the amount of genetic overlap is no more than what would be expected by chance, indicating that different stem cells have largely different gene expression patterns. Taken together, these studies identify many genes not previously associated with neural progenitor cell biology and also provide a rational scheme for stratification of microarray data for functional analysis.


Subject(s)
Brain/metabolism , Embryo, Mammalian/embryology , Gene Expression Profiling , Neurons/cytology , Stem Cells/metabolism , Animals , Brain/cytology , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Hematopoietic Stem Cells/metabolism , In Situ Hybridization , Mice , Oligonucleotide Array Sequence Analysis
14.
Dev Neurosci ; 25(2-4): 162-72, 2003.
Article in English | MEDLINE | ID: mdl-12966214

ABSTRACT

Central nervous system germinal zones contain stem cells that generate both neurons and glia. In the recent past, these cells have been isolated, maintained in a variety of culture systems and used in vitro for subsequent characterization of molecular mechanisms underlying brain development. Factors that govern cell fate choices of these neural stem cells have not been fully elucidated, but recent studies suggest that age at the time of culture is an important intrinsic mechanism. Stem cell mitogens and Notch-DSL signaling are significant extrinsic factors. In the current study, we compare neurosphere cultures propagated from animals on embryonic day 12, embryonic day 18 and the day of birth and stimulated to divide by either basic fibroblast growth factor (bFGF) or transforming growth factor-alpha (TGF-alpha). As described for other systems, when bFGF was used, clonal neurospheres derived from the youngest age gave rise to a greater percentage of neurons. When TGF-alpha, acting via the epidermal growth factor receptor, was used, this effect was not observed, with neurospheres from younger animals giving rise to a similar percentage of neurons as those derived from older animals suggesting that this growth factor was either stimulating a different population of stem cells to proliferate, or that it was capable of overriding intrinsic mechanisms. Other differences were also observed when the two growth factors were compared, including age-dependent differences in the numbers of putative astrocytes and oligodendrocytes formed. We further assessed age-dependent influences on cell fate by assessing the effects of a lentivirally transduced constitutively activated Notch receptor on cell fate. At all ages studied, Notch activation resulted in a significantly greater number of GFAP-positive cells, seemingly overriding the greater neurogenic potential of younger stem cells. These data suggest that both extrinsic and intrinsic factors differentially regulate cell fate choices of progenitors during cortical development.


Subject(s)
Aging/physiology , Cell Differentiation/physiology , Cerebral Cortex/embryology , Neurons/cytology , Stem Cells/cytology , Animals , Astrocytes/cytology , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Embryo, Mammalian , Embryonic and Fetal Development/physiology , Fibroblast Growth Factor 2/pharmacology , Fluorescent Antibody Technique , Genetic Vectors , Humans , Lentivirus/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neurons/drug effects , Receptors, Notch , Stem Cells/drug effects , Transduction, Genetic , Transforming Growth Factor alpha/pharmacology
15.
J Neurosci ; 23(3): 748-57, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12574403

ABSTRACT

RAS proteins are critical regulators of mitosis and are mutationally activated in many human tumors. RAS signaling is also known to mediate long-term potentiation (LTP) and long-term memory formation in postmitotic neurons, in part through activation of the RAF-MEK-ERK pathway. The RAS effector RIN1 appears to function through competitive inhibition of RAS-RAF binding and also through diversion of RAS signaling to alternate pathways. We show that RIN1 is preferentially expressed in postnatal forebrain neurons in which it is localized in dendrites and physically associated with RAS, suggesting a role in RAS-mediated postsynaptic neuronal plasticity. Mice with an Rin1 gene disruption showed a striking enhancement in amygdala LTP. In addition, two independent behavioral tests demonstrated elevated amygdala-dependent aversive memory in Rin1(-/-) mice. These results indicate that RIN1 serves as an inhibitory modulator of neuronal plasticity in aversive memory formation.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Long-Term Potentiation/physiology , Memory/physiology , Neuronal Plasticity/physiology , rab GTP-Binding Proteins , ras Proteins/metabolism , Amygdala/physiology , Animals , Carrier Proteins/genetics , Cell Count , Conditioning, Classical , Dendrites/metabolism , Fear/physiology , Fetal Viability/genetics , Hippocampus/physiology , Humans , Learning/physiology , Long-Term Potentiation/genetics , Maze Learning/physiology , Mice , Mice, Knockout , Neurons/metabolism , Neurons/ultrastructure , Prosencephalon/chemistry , Prosencephalon/cytology , Prosencephalon/metabolism , Protein Binding/physiology
SELECTION OF CITATIONS
SEARCH DETAIL