Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Alzheimers Dement (N Y) ; 6(1): e12093, 2020.
Article in English | MEDLINE | ID: mdl-33083515

ABSTRACT

INTRODUCTION: Gln-1062 (MEMOGAIN) is an intranasally administered lipophilic prodrug of galantamine. Based on high brain-to-blood concentrations observed in pre-clinical studies, Gln-1062 is expected to have superior cognitive efficacy compared to oral galantamine. METHODS: Forty-eight healthy elderly subjects were randomized 12:4 to Gln-1062 (5.5, 11, or 22 mg, b.i.d., for 7 days) or placebo. Safety, tolerability, pharmacokinetics, and pharmacodynamics were assessed repeatedly. Pharmacokinetics were compared with 16 mg oral galantamine. RESULTS: Gln-1062 up to 22 mg, b.i.d., was well tolerated. Gln-1062 plasma concentrations increased immediately following dosing (median Tmax of 0.5 hour [range 0.5-1.0]). Cmax and AUC0-last increased in a dose-linear manner over all three dose levels. Gln-1062 was rapidly cleaved into galantamine. Gln-1062 significantly improved adaptive tracking (sustained attention) with 1.95% (95% confidence interval [CI] 0.630-3.279, P = 0.0055) compared to placebo after correction for individual baseline performance. DISCUSSION: Gln-1062 was considered to be safe and caused fewer gastrointestinal side effects than oral galantamine. Gln-1062 behaved pharmacokinetically as expected and improved performance on cognitive tests.

2.
PLoS One ; 12(8): e0182896, 2017.
Article in English | MEDLINE | ID: mdl-28837568

ABSTRACT

Progranulin (PGRN) is a multifunctional protein that is widely expressed throughout the brain, where it has been shown to act as a critical regulator of CNS inflammation and also functions as an autocrine neuronal growth factor, important for long-term neuronal survival. PGRN has been shown to activate cell signaling pathways regulating excitoxicity, oxidative stress, and synaptogenesis, as well as amyloidogenesis. Together, these critical roles in the CNS suggest that PGRN has the potential to be an important therapeutic target for the treatment of various neurodegenerative disorders, particularly Alzheimer's disease (AD). AD is the leading cause of dementia and is marked by the appearance of extracellular plaques consisting of aggregates of amyloid-ß (Aß), as well as neuroinflammation, oxidative stress, neuronal loss and synaptic atrophy. The ability of PGRN to target multiple key features of AD pathophysiology suggests that enhancing its expression may benefit this disease. Here, we describe the application of PGRN gene transfer using in vivo delivery of lentiviral expression vectors in a transgenic mouse model of AD. Viral vector delivery of the PGRN gene effectively enhanced PGRN expression in the hippocampus of Tg2576 mice. This elevated PGRN expression significantly reduced amyloid plaque burden in these mice, accompanied by reductions in markers of inflammation and synaptic atrophy. The overexpression of PGRN was also found to increase activity of neprilysin, a key amyloid beta degrading enzyme. PGRN regulation of neprilysin activity could play a major role in the observed alterations in plaque burden. Thus, PGRN may be an effective therapeutic target for the treatment of AD.


Subject(s)
Alzheimer Disease/therapy , Disease Models, Animal , Genetic Therapy , Intercellular Signaling Peptides and Proteins/genetics , Plaque, Atherosclerotic , Synapses/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Sequence , Animals , Female , Genetic Vectors , Granulins , Hippocampus/metabolism , Hippocampus/pathology , Intercellular Signaling Peptides and Proteins/administration & dosage , Lentivirus/genetics , Mice , Mice, Transgenic , Progranulins
3.
PLoS One ; 12(3): e0174784, 2017.
Article in English | MEDLINE | ID: mdl-28358904

ABSTRACT

Progranulin (PGRN) is a glycoprotein with multiple roles in normal and disease states. Mutations within the GRN gene cause frontotemporal lobar degeneration (FTLD). The affected neurons display distinctive TAR DNA binding protein 43 (TDP-43) inclusions. How partial loss of PGRN causes TDP-43 neuropathology is poorly understood. TDP-43 inclusions are also found in affected neurons of patients with other neurodegenerative diseases including amyotrophic lateral sclerosis (ALS) and Alzheimer's disease. In ALS, TDP-43 inclusions are typically also immunoreactive for fused in sarcoma (FUS). Mutations within TDP-43 or FUS are themselves neuropathogenic in ALS and some cases of FTLD. We used the outgrowth of caudal primary motor neurons (MNs) in zebrafish embryos to investigate the interaction of PGRN with TDP-43 and FUS in vivo. As reported previously, depletion of zebrafish PGRN-A (zfPGRN-A) is associated with truncated primary MNs and impaired motor function. Here we found that depletion of zfPGRN-A results in primary MNs outgrowth stalling at the horizontal myoseptum, a line of demarcation separating the myotome into dorsal and ventral compartments that is where the final destination of primary motor is assigned. Successful axonal outgrowth beyond the horizontal myoseptum depends in part upon formation of acetylcholine receptor clusters and this was found to be disorganized upon depletion of zfPGRN-A. PGRN reversed the effects of zfPGRN-A knockdown, but a related gene, zfPGRN-1, was without effect. Both knockdown of TDP-43 or FUS, as well as expression of humanTDP-43 and FUS mutants results in MN abnormalities that are reversed by co-expression of hPGRN mRNA. Neither TDP-43 nor FUS reversed MN phenotypes caused by the depletion of PGRN. Thus TDP-43 and FUS lie upstream of PGRN in a gene complementation pathway. The ability of PGRN to override TDP-43 and FUS neurotoxicity due to partial loss of function or mutation in the corresponding genes may have therapeutic relevance.


Subject(s)
DNA-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Motor Neurons/metabolism , RNA-Binding Protein FUS/metabolism , Zebrafish Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mutation/genetics , RNA-Binding Protein FUS/genetics , Zebrafish , Zebrafish Proteins/genetics
4.
Alzheimers Dement (N Y) ; 2(1): 13-22, 2016 Jan.
Article in English | MEDLINE | ID: mdl-29067291

ABSTRACT

INTRODUCTION: Gln-1062 (Memogain) is a pharmacologically inactive prodrug of galantamine. Owing to its lipophilic nature, it preferentially enters the brain, where it is cleaved into active galantamine. Gln-1062 is expected to have fewer peripheral side effects than other cholinesterase inhibitors, with improved effectiveness. METHODS: This was a double-blind, comparator and placebo-controlled, sequential cohort, single ascending dose study in 58 healthy subjects with Gln-1062 in doses of 5.5, 11, 22, 33, and 44 mg, compared with oral galantamine 16 mg and donepezil 10 mg. Safety, tolerability, pharmacokinetics, and pharmacodynamics were assessed. RESULTS: Gln-1062 doses up to 33 mg were well tolerated and induced a dose-dependent increase in the plasma concentrations of Gln-1062 and galantamine. Gln-1062 had a dose-dependent positive effect on verbal memory and attention, mainly in the first hours after drug administration. DISCUSSION: Gln-1062 was better tolerated than galantamine in doses with the same molarity and led to improved effects in cognitive tests. This is most likely caused by the more favorable distribution ratio between peripheral and central cholinesterase inhibition. These results give reason for further exploration of this compound.

5.
PLoS One ; 10(10): e0139694, 2015.
Article in English | MEDLINE | ID: mdl-26439489

ABSTRACT

The development of effective neuroprotective therapies for Parkinson's disease (PD) has been severely hindered by the notable lack of an appropriate animal model for preclinical screening. Indeed, most models currently available are either acute in nature or fail to recapitulate all characteristic features of the disease. Here, we present a novel progressive model of PD, with behavioural and cellular features that closely approximate those observed in patients. Chronic exposure to dietary phytosterol glucosides has been found to be neurotoxic. When fed to rats, ß-sitosterol ß-d-glucoside (BSSG) triggers the progressive development of parkinsonism, with clinical signs and histopathology beginning to appear following cessation of exposure to the neurotoxic insult and continuing to develop over several months. Here, we characterize the progressive nature of this model, its non-motor features, the anatomical spread of synucleinopathy, and response to levodopa administration. In Sprague Dawley rats, chronic BSSG feeding for 4 months triggered the progressive development of a parkinsonian phenotype and pathological events that evolved slowly over time, with neuronal loss beginning only after toxin exposure was terminated. At approximately 3 months following initiation of BSSG exposure, animals displayed the early emergence of an olfactory deficit, in the absence of significant dopaminergic nigral cell loss or locomotor deficits. Locomotor deficits developed gradually over time, initially appearing as locomotor asymmetry and developing into akinesia/bradykinesia, which was reversed by levodopa treatment. Late-stage cognitive impairment was observed in the form of spatial working memory deficits, as assessed by the radial arm maze. In addition to the progressive loss of TH+ cells in the substantia nigra, the appearance of proteinase K-resistant intracellular α-synuclein aggregates was also observed to develop progressively, appearing first in the olfactory bulb, then the striatum, the substantia nigra and, finally, hippocampal and cortical regions. The slowly progressive nature of this model, together with its construct, face and predictive validity, make it ideal for the screening of potential neuroprotective therapies for the treatment of PD.


Subject(s)
Brain/pathology , Disease Models, Animal , Motor Activity/physiology , Neurons/pathology , Parkinson Disease, Secondary/pathology , Animals , Brain/metabolism , Brain/physiopathology , Neurons/metabolism , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/physiopathology , Rats , Rats, Sprague-Dawley , Sitosterols , alpha-Synuclein/metabolism
6.
PLoS One ; 9(5): e97032, 2014.
Article in English | MEDLINE | ID: mdl-24804730

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by tremor, rigidity and akinesia/bradykinesia resulting from the progressive loss of nigrostriatal dopaminergic neurons. To date, only symptomatic treatment is available for PD patients, with no effective means of slowing or stopping the progression of the disease. Progranulin (PGRN) is a 593 amino acid multifunction protein that is widely distributed throughout the CNS, localized primarily in neurons and microglia. PGRN has been demonstrated to be a potent regulator of neuroinflammation and also acts as an autocrine neurotrophic factor, important for long-term neuronal survival. Thus, enhancing PGRN expression may strengthen the cells resistance to disease. In the present study, we have used the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD to investigate the possible use of PGRN gene delivery as a therapy for the prevention or treatment of PD. Viral vector delivery of the PGRN gene was an effective means of elevating PGRN expression in nigrostriatal neurons. When PGRN expression was elevated in the SNC, nigrostriatal neurons were protected from MPTP toxicity in mice, along with a preservation of striatal dopamine content and turnover. Further, protection of nigrostriatal neurons by PGRN gene therapy was accompanied by reductions in markers of MPTP-induced inflammation and apoptosis as well as a complete preservation of locomotor function. We conclude that PGRN gene therapy may have beneficial effects in the treatment of PD.


Subject(s)
Dopaminergic Neurons/metabolism , Intercellular Signaling Peptides and Proteins/genetics , MPTP Poisoning/drug therapy , Parkinson Disease/genetics , Animals , Apoptosis/genetics , Disease Models, Animal , Dopamine/genetics , Dopamine/metabolism , Dopaminergic Neurons/pathology , Gene Transfer Techniques , Genetic Therapy , Granulins , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , MPTP Poisoning/genetics , MPTP Poisoning/pathology , Mice , Motor Activity/drug effects , Motor Activity/immunology , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Progranulins
7.
Exp Gerontol ; 50: 95-105, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24316034

ABSTRACT

Panax ginseng has been used in traditional Chinese medicine for centuries. Among its various benefits is a pluripotent targeting of the various events involved in neuronal cell death. This includes anti-inflammatory, anti-oxidant, and anti-apoptotic effects. Indeed, ginseng extract and its individual ginsenosides have been demonstrated to influence a number of biochemical markers implicated in Parkinson's disease (PD) pathogenesis. We have reported previously that administration of the ginseng extract, G115, afforded robust neuroprotection in two rodent models of PD. However, these traditional rodent models are acute in nature and do accurately recapitulate the progressive nature of the disease. Chronic exposure to the dietary phytosterol glucoside, ß-sitosterol ß-d-glucoside (BSSG) triggers the progressive development of neurological deficits, with behavioral and cellular features that closely approximate those observed in PD patients. Clinical signs and histopathology continue to develop for several months following cessation of exposure to the neurotoxic insult. Here, we utilized this model to further characterize the neuroprotective effects of the ginseng extract, G115. Oral administration of this extract significantly reduced dopaminergic cell loss, microgliosis, and accumulation of α-synuclein aggregates. Further, G115 administration fully prevented the development of locomotor deficits, in the form of reduced locomotor activity and coordination. These results suggest that ginseng extract may be a potential neuroprotective therapy for the treatment of PD.


Subject(s)
Neuroprotective Agents/therapeutic use , Panax , Parkinson Disease, Secondary/prevention & control , Phytotherapy/methods , Animals , Cell Death/drug effects , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical/methods , Encephalitis/chemically induced , Encephalitis/prevention & control , Female , Gait Disorders, Neurologic/chemically induced , Gait Disorders, Neurologic/prevention & control , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Plant Extracts/therapeutic use , Rats , Rats, Sprague-Dawley , Sitosterols , Substantia Nigra/pathology , alpha-Synuclein/metabolism
8.
Mol Neurodegener ; 5: 41, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-20946666

ABSTRACT

BACKGROUND: Progranulin (PGRN) encoded by the GRN gene, is a secreted glycoprotein growth factor that has been implicated in many physiological and pathophysiological processes. PGRN haploinsufficiency caused by autosomal dominant mutations within the GRN gene leads to progressive neuronal atrophy in the form of frontotemporal lobar degeneration (FTLD). This form of the disease is associated with neuronal inclusions that bear the ubiquitinated TAR DNA Binding Protein-43 (TDP-43) molecular signature (FTLD-U). The neurotrophic properties of PGRN in vitro have recently been reported but the role of PGRN in neurons is not well understood. Here we document the neuronal expression and functions of PGRN in spinal cord motoneuron (MN) maturation and branching in vivo using zebrafish, a well established model of vertebrate embryonic development. RESULTS: Whole-mount in situ hybridization and immunohistochemical analyses of zebrafish embryos revealed that zfPGRN-A is expressed within the peripheral and central nervous systems including the caudal primary (CaP) MNs within the spinal cord. Knockdown of zfPGRN-A mRNA translation mediated by antisense morpholino oligonucleotides disrupted normal CaP MN development resulting in both truncated MNs and inappropriate early branching. Ectopic over-expression of zfPGRN-A mRNA resulted in increased MN branching and rescued the truncation defects brought about by knockdown of zfPGRN-A expression. The ability of PGRN to interact with established MN developmental pathways was tested. PGRN over-expression was found to reverse the truncation defect resulting from knockdown of Survival of motor neuron 1 (smn1). This is involved in small ribonucleoprotein biogenesis RNA processing, mutations of which cause Spinal Muscular Atrophy (SMA) in humans. It did not reverse the MN defects caused by interfering with the neuronal guidance pathway by knockdown of expression of NRP-1, a semaphorin co-receptor. CONCLUSIONS: Expression of PGRN within MNs and the observed phenotypes resulting from mRNA knockdown and over-expression are consistent with a role in the regulation of spinal cord MN development and branching. This study presents the first in vivo demonstration of the neurotrophic properties of PGRN and suggests possible future therapeutic applications in the treatment of neurodegenerative diseases.

9.
BMC Neurosci ; 10: 130, 2009 Oct 27.
Article in English | MEDLINE | ID: mdl-19860916

ABSTRACT

BACKGROUND: Progranulin is a secreted high molecular weight growth factor bearing seven and one half copies of the cysteine-rich granulin-epithelin motif. While inappropriate over-expression of the progranulin gene has been associated with many cancers, haploinsufficiency leads to atrophy of the frontotemporal lobes and development of a form of dementia (frontotemporal lobar degeneration with ubiquitin positive inclusions, FTLD-U) associated with the formation of ubiquitinated inclusions. Recent reports indicate that progranulin has neurotrophic effects, which, if confirmed would make progranulin the only neuroprotective growth factor that has been associated genetically with a neurological disease in humans. Preliminary studies indicated high progranulin gene expression in spinal cord motor neurons. However, it is uncertain what the role of Progranulin is in normal or diseased motor neuron function. We have investigated progranulin gene expression and subcellular localization in cultured mouse embryonic motor neurons and examined the effect of progranulin over-expression and knockdown in the NSC-34 immortalized motor neuron cell line upon proliferation and survival. RESULTS: In situ hybridisation and immunohistochemical techniques revealed that the progranulin gene is highly expressed by motor neurons within the mouse spinal cord and in primary cultures of dissociated mouse embryonic spinal cord-dorsal root ganglia. Confocal microscopy coupled to immunocytochemistry together with the use of a progranulin-green fluorescent protein fusion construct revealed progranulin to be located within compartments of the secretory pathway including the Golgi apparatus. Stable transfection of the human progranulin gene into the NSC-34 motor neuron cell line stimulates the appearance of dendritic structures and provides sufficient trophic stimulus to survive serum deprivation for long periods (up to two months). This is mediated at least in part through an anti-apoptotic mechanism. Control cells, while expressing basal levels of progranulin do not survive in serum free conditions. Knockdown of progranulin expression using shRNA technology further reduced cell survival. CONCLUSION: Neurons are among the most long-lived cells in the body and are subject to low levels of toxic challenges throughout life. We have demonstrated that progranulin is abundantly expressed in motor neurons and is cytoprotective over prolonged periods when over-expressed in a neuronal cell line. This work highlights the importance of progranulin as neuroprotective growth factor and may represent a therapeutic target for neurodegenerative diseases including motor neuron disease.


Subject(s)
Cell Survival/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Motor Neurons/metabolism , Spinal Cord/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Line , Cell Proliferation , Cell Survival/genetics , Cells, Cultured , Cloning, Molecular , Fluorescent Antibody Technique , Ganglia, Spinal/metabolism , Ganglia, Spinal/ultrastructure , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Granulins , Humans , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Microscopy, Confocal , Motor Neurons/ultrastructure , Progranulins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/ultrastructure , Transfection
10.
Am J Pathol ; 170(1): 399-415, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17200211

ABSTRACT

We previously reported that truncation of Notch1 (N1) by provirus insertion leads to overexpression of both the intracellular (N1(IC)) and the extracellular (N1(EC)) domains. We produced transgenic (Tg) mice expressing N1(EC) in T cells and in cells of the myeloid lineage under the regulation of the CD4 gene. These CD4C/N1(EC) Tg mice developed vascular disease, predominantly in the liver: superficial distorted vessels, cavernae, lower branching of parenchymal vessels, capillarized sinusoids, and aberrant smooth muscle/endothelial cell topography. The disease developed in lethally irradiated normal mice transplanted with Tg bone marrow or fetal liver cells as well as in Rag-/- Tg mice. In nude mice transplanted with fetal liver cells from (ROSA26 x CD4C/N1(EC)) F1 Tg mice, abnormal vessels were of recipient origin. Transplantation of Tg peritoneal macrophages into normal recipients also induced abnormal vessels. These Tg macrophages showed impaired functions, and their conditioned medium inhibited the proliferation of liver sinusoid endothelial cells in vitro. The Egr-1 gene and some of its targets (Jag1, FIII, FXIII-A, MCP-1, and MCP-5), previously implicated in hemangioma or vascular malformations, were overexpressed in Tg macrophages. These results show that myeloid cells can be reprogrammed by N1(EC) to induce vascular malformations through a paracrine pathway.


Subject(s)
Liver Diseases/genetics , Myeloid Cells/physiology , Peripheral Vascular Diseases/genetics , Receptor, Notch1/genetics , Animals , Fetal Tissue Transplantation , Liver/blood supply , Liver/pathology , Liver Diseases/pathology , Liver Transplantation , Macrophages, Peritoneal/physiology , Macrophages, Peritoneal/transplantation , Mice , Mice, Nude , Mice, Transgenic , Paracrine Communication , Peripheral Vascular Diseases/pathology , Protein Structure, Tertiary/genetics , Receptor, Notch1/biosynthesis , Up-Regulation
11.
Dev Biol ; 300(2): 497-511, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17046738

ABSTRACT

The Notch signaling pathway is involved in a variety of developmental processes. Here, we characterize the phenotypes developing in the reproductive organs of male transgenic (Tg) mice constitutively expressing the activated mouse Notch1 intracellular domain (Notch1(intra)) under the regulatory control of the mouse mammary tumor virus (MMTV) long terminal repeat (LTR). Tg expression was detected in testis, vas deferens and epididymis by Northern blot analysis. In situ hybridization with a Notch1-specific probe lacked sensitivity to detect expression in normal-appearing cells, but demonstrated expression in hyperplastic epithelial cells of the vas deferens, epididymis and efferent ducts. Tg males from three independent founder lines were sterile. Histological analysis of reproductive organs of young Tg males (postnatal ages 8 and 21) showed no difference compared to those of non-Tg males. In contrast, in adult Tg mice from day 38 onwards, the efferent ducts, the vas deferens and most epididymal segments revealed bilateral epithelial cell hyperplasia with absence of fully differentiated epithelial cells. Electron microscopy confirmed the uniformly undifferentiated state of these cells. Immunohistochemistry with anti-PCNA antibody also revealed enhanced proliferation of Tg epididymis. In adult Tg testis, the different generations of germ cells of seminiferous tubules appeared normal, although some tubules were highly dilated and revealed an absence of early and/or late spermatids. The epithelial cells of the Tg tubuli recti and rete testis were not abnormal, but the rete testis was highly dilated and contained numerous spermatozoa, suggesting a downstream blockage. Consistent with a blockage of efferent ducts often seen at the rete testis/efferent duct interface, spermatozoa were absent in epididymis of all adult Tg mice and in all highly hyperplastic efferent duct tubules of these Tg mice. Such a blockage was visualized by injection of Evans blue dye into the rete testis lumen. Finally, the presence of ectopic hyperplastic efferent duct tubules was observed within the testicular parenchyma itself, outside their normal territory, suggesting that Notch1 signaling is involved in the establishment of these borders. This phenotype seems to represent a novel developmental defect in mammals. Together, these results show that constitutive Notch1 signaling significantly affects the development of male reproductive organs.


Subject(s)
Epididymis/pathology , Infertility, Male/genetics , Infertility, Male/pathology , Receptor, Notch1/biosynthesis , Receptor, Notch1/genetics , Vas Deferens/pathology , Animals , Epididymis/growth & development , Female , Humans , Hyperplasia , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Notch1/physiology , Signal Transduction/genetics , Testis/pathology
12.
J Immunol ; 177(4): 2153-66, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16887975

ABSTRACT

We previously found that provirus insertion in T cell tumors of mouse mammary tumor virus/c-myc transgenic (Tg) mice induced two forms of Notch1 mutations. Type I mutations generated two truncated molecules, one intracellular (IC) (Notch1(IC)) and one extracellular (Notch1(EC)), while in type II mutations Notch1 was deleted of its C terminus (Notch1(DeltaCT)). We expressed these mutants in Tg mice using the CD4 promoter. Both Notch1(IC) and Notch1(DeltaCT), but not Notch1(EC), Tg mice developed double-positive (DP) thymomas. These disseminated more frequently in Notch1(DeltaCT) Tg mice. Double (Notch1(IC) x myc) or (Notch1(DeltaCT) x myc) Tg mice developed thymoma with a much shorter latency than single Tg mice, providing genetic evidence of a collaboration between these two oncogenes. FACS analysis of preleukemic thymocytes did not reveal major T cell differentiation anomalies, except for a higher number of DP cells and an accumulation of TCR(high)CD2(high)CD25(high) DP cells in Notch1(IC), and less so in Notch1(DeltaCT) Tg mice. This was associated with enhanced in vivo thymocyte proliferation. However, Notch1(IC), but not Notch1(DeltaCT), DP thymocytes were protected against apoptosis induced in vivo by dexamethasone and anti-CD3 and in vitro by anti-CD3/CD28 Abs. This indicates that the C terminus of Notch1 and/or the conserved regulation by its ligands have a significant impact on the induced T cell phenotype. Therefore, Notch1(IC) and Notch1(DeltaCT) behave as oncogenes for T cells. Because these two Notch1 mutations are very similar to those described in some forms of human T cell leukemia, these Tg mice may represent relevant models of these human leukemias.


Subject(s)
Apoptosis/genetics , Cell Differentiation/genetics , Gene Deletion , Leukemia-Lymphoma, Adult T-Cell/genetics , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Thymus Gland/immunology , Thymus Neoplasms/secondary , Animals , Apoptosis/immunology , Cell Differentiation/immunology , Disease Models, Animal , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukemia-Lymphoma, Adult T-Cell/pathology , Ligands , Lymphatic Metastasis , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Preleukemia/genetics , Preleukemia/immunology , Preleukemia/pathology , Protein Binding/immunology , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Receptor, Notch1/biosynthesis , Receptor, Notch1/chemistry , Thymoma/genetics , Thymoma/immunology , Thymoma/pathology , Thymoma/secondary , Thymus Gland/pathology , Thymus Neoplasms/genetics , Thymus Neoplasms/immunology , Thymus Neoplasms/pathology
13.
J Virol ; 79(10): 6377-91, 2005 May.
Article in English | MEDLINE | ID: mdl-15858021

ABSTRACT

CD4(+)- and CD8(+)-T-cell death is a frequent immunological dysfunction associated with the development of human AIDS. We studied a murine model of AIDS, the CD4C/HIV transgenic (Tg) mouse model, to assess the importance of the apoptotic pathway in human immunodeficiency virus type 1 (HIV-1) pathogenesis. In these Tg mice, Nef is the major determinant of the disease and is expressed in immature and mature CD4(+) T cells and in cells of the macrophage/myeloid lineage. We report here a novel AIDS-like phenotype: enhanced death, most likely by apoptosis (as assessed by 7-aminoactinomycin D and annexin V/propidium iodide staining), of Tg thymic and peripheral CD4(+) and CD8(+) T cells. The Tg CD4(+) and CD8(+) T cells were also more susceptible to cell death after activation in vitro in mixed lymph node (LN) cultures. However, activation-induced cell death was not higher in Tg than in non-Tg-purified CD4(+) T cells. In addition, expression of Fas and FasL, assessed by flow cytometry, was increased in CD4(+) and CD8(+) T cells from Tg mice compared to that of non-Tg littermates. Despite the enhanced expression of Fas and FasL on Tg CD4(+) and CD8(+) T cells, Fas (lpr/lpr) and FasL (gld/gld) mutant CD4C/HIV Tg mice developed an AIDS-like disease indistinguishable from lpr/+ and gld/+ CD4C/HIV Tg mice, including loss of CD4(+) T cells. Similarly, CD4C/HIV Tg mice homozygous for mutations of two other genes implicated in cell death (interleukin-1beta-converting enzyme [ICE], tumor necrosis factor receptor 1 [TNFR-1]) developed similar AIDS-like disease as their respective heterozygous controls. Moreover, the double-Tg mice from a cross between the Bcl2/Wehi25 and CD4C/HIV Tg mice showed no major protection against disease. These results represent genetic evidence for the dispensable role of Fas, FasL, ICE, and TNFR-1 on the development of both T-cell loss and organ disease of these Tg mice. They also provide compelling evidence on the lack of protection by Bcl2 against Tg CD4(+)-T-cell death. In view of the high resemblance between numerous phenotypes observed in the CD4C/HIV Tg mice and in human AIDS, our findings are likely to be relevant for the human disease.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , Apoptosis , Caspase 1/deficiency , Genes, nef/physiology , HIV-1/genetics , Membrane Glycoproteins/metabolism , Proto-Oncogene Proteins/metabolism , T-Lymphocytes/physiology , TNF Receptor-Associated Factor 1/deficiency , fas Receptor/metabolism , Acquired Immunodeficiency Syndrome/genetics , Acquired Immunodeficiency Syndrome/pathology , Animals , Caspase 1/genetics , Disease Models, Animal , Edema/pathology , Fas Ligand Protein , Kidney/pathology , Lymph Nodes/pathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2 , T-Lymphocytes/immunology , TNF Receptor-Associated Factor 1/genetics , Thymus Gland/pathology , Weight Loss , fas Receptor/genetics
14.
Virology ; 327(2): 273-86, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15351215

ABSTRACT

HIV-1 Nef is a critical determinant of pathogenicity in humans and transgenic (Tg) mice. To gain a better understanding of the molecular mechanisms by which Nef induces an AIDS-like disease in Tg mice, a mutational analysis of the N-terminal domain, involved in anchoring Nef to the plasma membrane, was carried out. The pathogenic effects of these Nef mutant alleles were evaluated in Tg mice by FACS analysis and by histopathological assessment. Mutation of the myristoylation site (G2A) completely abrogated the development of the AIDS-like organ disease in Tg mice, although partial downregulation of the CD4 cell surface protein and depletion of peripheral CD4+ T-cells, but not of CD4(+)CD8+ thymocytes, still occurred. Despite that, the peripheral CD4+ T cells expressing Nef(G2A) show normal spontaneous proliferation in vivo or after stimulation in vitro, including in an allogenic mixed leukocyte reaction (MLR). Three other internal deletion mutants of Nef, spanning amino acids 8-17 (Nef(Delta8-17)), 25-35 (Nef(Delta25-35)), and 57-66 (Nef(Delta57-66)), were also studied. Nef(Delta8-17) retained full pathogenic potential, although Nef(Delta25-35) and Nef(Delta57-66) Tg mice were free of organ disease. However, Nef(Delta25-35) Tg mice exhibited disorganization of thymic architecture and a partial depletion of peripheral CD4+ T cells. These data indicate that myristoylation and other regions at the N-terminus of Nef (aa 25-35 and 57-66) are involved in mediating severe T-cell phenotypes and organ disease, although residues 8-17 are dispensable for these Nef functions. In addition, these results indicate that at least some of the CD4+ T-cell phenotypes can develop independently of the other AIDS-like organ phenotypes. This apparent segregation of different Nef-mediated phenotypes suggests distinct mechanisms of Nef action in different populations of target cells, and may be relevant to human AIDS.


Subject(s)
DNA Mutational Analysis/methods , Gene Products, nef/chemistry , Gene Products, nef/genetics , HIV Infections/pathology , HIV-1/pathogenicity , Animals , Down-Regulation , Flow Cytometry , Gene Products, nef/metabolism , HIV Infections/mortality , HIV Infections/virology , Humans , Lymphocyte Activation , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Myristic Acid/metabolism , Sequence Deletion , Spleen/cytology , Spleen/immunology , Thymus Gland/cytology , Thymus Gland/immunology , nef Gene Products, Human Immunodeficiency Virus
15.
J Virol ; 78(10): 5244-57, 2004 May.
Article in English | MEDLINE | ID: mdl-15113906

ABSTRACT

The cellular and molecular mechanisms of dysfunction and depletion of CD4+ T lymphocytes over the course of human immunodeficiency virus type 1 (HIV-1) infection are still incompletely understood, but chronic immune activation is thought to play an important role in disease progression. We studied CD4+ T-cell biology in CD4C/HIV transgenic (Tg) mice, in which Nef expression is sufficient to induce a severe AIDS-like disease including a preferential decrease of CD4+ T cells. We show here that Nef-expressing Tg CD4+ T cells exhibit an activated/memory-like phenotype which appears to be independent of antigenic stimulation, as documented in experiments involving breeding with AD10 TcR Tg mice. In addition, in vivo bromodeoxyuridine incorporation showed that a larger proportion of Tg than non-Tg CD4+ T cells entered the S phase. However, in vitro, Tg CD4+ T cells were found to have a very limited capacity to divide in response to stimulation with anti-CD3 and anti-CD28 or in allogeneic mixed leukocyte reactions. Interestingly, despite these observations, the deletion of Tg CD4+ T cells had little impact on the development of other AIDS-like organ phenotypes. Thus, the Nef-induced chronic activation of CD4+ T cells may exhaust the T-cell pool and may contribute to the thymic atrophy and the low number of CD4+ T cells observed in these Tg mice.


Subject(s)
Acquired Immunodeficiency Syndrome/etiology , CD4-Positive T-Lymphocytes/immunology , Gene Products, nef/physiology , HIV-1/immunology , Lymphocyte Activation , Animals , Bromodeoxyuridine/metabolism , Immunologic Memory , Mice , Mice, Transgenic , nef Gene Products, Human Immunodeficiency Virus
16.
J Virol ; 77(24): 13161-70, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14645573

ABSTRACT

Some murine leukemia viruses (MuLVs), among them Cas-Br-E and ts-1 MuLVs, are neurovirulent, inducing spongiform myeloencephalopathy and hind limb paralysis in susceptible mice. It has been shown that the env gene of these viruses harbors the determinant of neurovirulence. It appears that neuronal loss occurs by an indirect mechanism, since the target motor neurons have not been found to be infected. However, the pathogenesis of the disease remains unclear. Several lymphokines, cytokines, and other cellular effectors have been found to be aberrantly expressed in the brains of infected mice, but whether these are required for the development of the neurodegenerative lesions is not known. In an effort to identify the specific effectors which are indeed required for the initiation and/or development of spongiform myeloencephalopathy, we inoculated gene-deficient (knockout [KO]) mice with ts-1 MuLV. We show here that interleukin-6 (IL-6), inducible nitric oxide synthetase (iNOS), ICE, Fas, Fas ligand (FasL), and TNF-R1 KO mice still develop signs of disease. However, transgenic mice overexpressing Bcl-2 in neurons (NSE/Bcl-2) were largely protected from hind limb paralysis and had less-severe spongiform lesions. These results indicate that motor neuron death occurs in this disease at least in part by a Bcl-2-inhibitable pathway not requiring the ICE, iNOS, Fas/FasL, TNF-R1, and IL-6 gene products.


Subject(s)
Central Nervous System Viral Diseases/prevention & control , Central Nervous System Viral Diseases/virology , Leukemia Virus, Murine/pathogenicity , Proto-Oncogene Proteins c-bcl-2/metabolism , Retroviridae Infections/virology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Caspase 1/genetics , Caspase 1/metabolism , Central Nervous System Viral Diseases/metabolism , Fas Ligand Protein , Interleukin-6/genetics , Interleukin-6/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C3H/metabolism , Mice, Knockout , Mice, Transgenic , Nerve Degeneration/prevention & control , Nerve Degeneration/virology , Neurons/metabolism , Neurons/pathology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Retroviridae Infections/metabolism , Retroviridae Infections/prevention & control , fas Receptor/genetics , fas Receptor/metabolism
17.
J Virol ; 77(21): 11733-44, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14557658

ABSTRACT

CD4C/human immunodeficiency virus (HIV) transgenic mice develop an AIDS-like disease. We used this model to study the effects of HIV-1 on dendritic cells (DC). We found a progressive decrease in total DC numbers in the lymph nodes, with a significant accumulation of CD11b(Hi) DC. In the thymus, the recovery of transgenic CD8alpha(+) DC had a tendency to be lower. Spleen DC were augmented in the marginal zone. Transgenic DC showed a decreased capacity to present antigen in vitro, consistent with their reduced major histocompatibility complex class II expression and impaired maturation profile. The accumulation of immature DC may contribute to disease and may reflect an adaptive advantage for the virus by favoring its replication and preventing the generation of fully functional antiviral responses.


Subject(s)
CD11b Antigen/analysis , Dendritic Cells/immunology , HIV Infections/immunology , HIV-1/physiology , Animals , Antigen-Presenting Cells/immunology , Bone Marrow Cells/immunology , CD4 Antigens/genetics , CD4 Antigens/metabolism , Dendritic Cells/virology , HIV Infections/pathology , HIV Infections/virology , HIV-1/genetics , Humans , Lymph Nodes/immunology , Mice , Mice, Transgenic , Spleen/cytology , Spleen/immunology , Transgenes
18.
J Virol ; 77(21): 11745-53, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14557659

ABSTRACT

Vacuolar myelopathy (VM) is a frequent central nervous system complication of human immunodeficiency virus type 1 (HIV-1) infection. We report here that transgenic (Tg) mice expressing even low levels of Nef in oligodendrocytes under the regulation of the myelin basic protein (MBP) promoter (MBP/HIV(Nef)) developed VM similar to the human disease in its appearance and topography. The spinal cords of these Tg mice showed lower levels of the myelin proteins MAG and CNPase and of the 21-kDa isoform of MBP prior to the development of vacuoles. In addition, Tg oligodendrocytes in primary in vitro cultures appeared morphologically more mature but, paradoxically, exhibited a less mature phenotype based on O4, O1, CNPase, and MBP staining. In particular, mature CNPase(+) MBP(+) Tg oligodendrocytes were less numerous than non-Tg oligodendrocytes. Therefore, Nef appears to affect the proper differentiation of oligodendrocytes. These data suggest that even low levels of Nef expression in human oligodendrocytes may be responsible for the development of VM in HIV-1-infected individuals.


Subject(s)
Gene Products, nef/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Vacuoles/ultrastructure , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Animals , Cell Differentiation , Cells, Cultured , Gene Products, nef/genetics , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin-Associated Glycoprotein/metabolism , Phenotype , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Diseases/pathology , Spinal Cord Diseases/virology , Transgenes
19.
Am J Pathol ; 161(1): 321-35, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12107117

ABSTRACT

We previously reported that a severe acquired immune deficiency syndrome-like disease develops in transgenic (Tg) mice expressing the human immunodeficiency virus-1 in its natural target cells: immature and mature CD4(+) T cells and cells of the macrophage/dendritic lineage. Here, we show that these mice also develop cardiac disease, characterized most prominently by a focal myocytolysis, occasionally by myocarditis and by deposition of endogenous immunoglobulin on cardiomyocytes. Microfil perfusion demonstrated widespread coronary arteriospasm and echocardiographic analysis revealed depressed cardiac function in Tg mice. A higher (but still modest) level of cardiomyocyte apoptosis was detected in Tg as compared to non-Tg hearts. Tg expression was detected in some of the infiltrating mononuclear cells, but not in cardiomyocytes or in cells of the heart vessels, suggesting a human immunodeficiency virus-1-induced disease process mediated by cells of the immune system. The similarity of the heart disease observed in these Tg mice to that observed in acquired immune deficiency syndrome patients suggests a common pathogenesis.


Subject(s)
Cardiomyopathies/etiology , Gene Products, nef/metabolism , HIV-1/metabolism , Immune System/metabolism , Mice, Transgenic/metabolism , Animals , Apoptosis , CD4 Antigens/genetics , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Coronary Vessels/physiopathology , Cytoskeletal Proteins/metabolism , Dystroglycans , HIV-1/genetics , Heart/physiopathology , Immunoglobulins/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic/genetics , Mutation , Myocardium/metabolism , nef Gene Products, Human Immunodeficiency Virus
20.
J Infect Dis ; 185(8): 1103-14, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11930320

ABSTRACT

The availability of CD4C/HIV(MutA) transgenic (Tg) mice expressing human immunodeficiency virus type 1 in immune cells and developing an AIDS-like disease has provided the opportunity to devise a model of mucosal candidiasis that closely mimics the clinical and pathologic features of candidal infection in human AIDS. After intraoral infection with Candida albicans, oral burdens were strikingly elevated in the Tg mice, compared with non-Tg littermates (P<.05), during primary infection, a 6-10-week carrier state, and a marked terminal outgrowth preceding death. The chronic carrier state was absent in the non-Tg mice because of clearing of C. albicans. Candida hyphae penetrated the epithelium of the oral cavity, esophagus, and cardial-atrium fold of the stomach, accompanied by a mononuclear cell infiltrate. Immunohistochemical analysis suggested that decreased frequencies of major histocompatibility complex class II-expressing cells, combined with reduced CD4+ cells, may underlie the susceptibility to mucosal candidiasis in these Tg mice.


Subject(s)
Candidiasis, Oral/etiology , HIV-1/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , Candidiasis, Oral/pathology , Disease Models, Animal , HIV Infections/complications , Histocompatibility Antigens Class II/analysis , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C3H , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...