Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Cureus ; 16(6): e62224, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39006590

ABSTRACT

Colonic xanthomas are a rare finding, particularly when combined with a tubular adenoma in a single polyp. While transformation to malignancy is not thought to be higher than that of a tubular adenoma alone, there is still concern as to the pathophysiology of xanthoma formation within the colon and what that may mean for patient outcomes. Here, we present a patient undergoing a routine screening colonoscopy with the removal of a rectosigmoid polyp consistent with xanthoma and tubular adenoma histopathology. Proper follow-up for identification of possible metabolic derangements and increased colonic surveillance is recommended to mitigate the risk of further xanthoma or adenocarcinoma formation.

2.
Front Cell Dev Biol ; 7: 201, 2019.
Article in English | MEDLINE | ID: mdl-31612134

ABSTRACT

Homology between mitochondrial DNA (mtDNA) and nuclear DNA of mitochondrial origin (nuMTs) causes confounding when aligning short sequence reads to the reference human genome, as the true sequence origin cannot be determined. Using a systematic in silico approach, we here report the impact of all potential mitochondrial variants on alignment accuracy and variant calling. A total of 49,707 possible mutations were introduced across the 16,569 bp reference mitochondrial genome (16,569 × 3 alternative alleles), one variant at-at-time. The resulting in silico fragmentation and alignment to the entire reference genome (GRCh38) revealed preferential mapping of mutated mitochondrial fragments to nuclear loci, as variants increased loci similarity to nuMTs, for a total of 807, 362, and 41 variants at 333, 144, and 27 positions when using 100, 150, and 300 bp single-end fragments. We subsequently modeled these affected variants at 50% heteroplasmy and carried out variant calling, observing bias in the reported allele frequencies in favor of the reference allele. Four variants (chrM:6023A, chrM:4456T, chrM:5147A, and chrM:7521A) including a possible hypertension factor, chrM:4456T, caused 100% loss of coverage at the mutated position (with all 100 bp single-end fragments aligning to homologous, nuclear positions instead of chrM), rendering these variants undetectable when aligning to the entire reference genome. Furthermore, four mitochondrial variants reported to be pathogenic were found to cause significant loss of coverage and select haplogroup-defining SNPs were shown to exacerbate the loss of coverage caused by surrounding variants. Increased fragment length and use of paired-end reads both improved alignment accuracy.

4.
Neurobiol Dis ; 76: 46-56, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25662335

ABSTRACT

Huntington disease (HD) is an inherited, fatal neurodegenerative disease with no disease-modifying therapy currently available. In addition to characteristic motor deficits and atrophy of the caudate nucleus, signature hallmarks of HD include behavioral abnormalities, immune activation, and cortical and white matter loss. The identification and validation of novel therapeutic targets that contribute to these degenerative cellular processes may lead to new interventions that slow or even halt the course of this insidious disease. Semaphorin 4D (SEMA4D) is a transmembrane signaling molecule that modulates a variety of processes central to neuroinflammation and neurodegeneration including glial cell activation, neuronal growth cone collapse and apoptosis of neural precursors, as well as inhibition of oligodendrocyte migration, differentiation and process formation. Therefore, inhibition of SEMA4D signaling could reduce CNS inflammation, increase neuronal outgrowth and enhance oligodendrocyte maturation, which may be of therapeutic benefit in the treatment of several neurodegenerative diseases, including HD. To that end, we evaluated the preclinical therapeutic efficacy of an anti-SEMA4D monoclonal antibody, which prevents the interaction between SEMA4D and its receptors, in the YAC128 transgenic HD mouse model. Anti-SEMA4D treatment ameliorated neuropathological signatures, including striatal atrophy, cortical atrophy, and corpus callosum atrophy and prevented testicular degeneration in YAC128 mice. In parallel, a subset of behavioral symptoms was improved in anti-SEMA4D treated YAC128 mice, including reduced anxiety-like behavior and rescue of cognitive deficits. There was, however, no discernible effect on motor deficits. The preservation of brain gray and white matter and improvement in behavioral measures in YAC128 mice treated with anti-SEMA4D suggest that this approach could represent a viable therapeutic strategy for the treatment of HD. Importantly, this work provides in vivo demonstration that inhibition of pathways initiated by SEMA4D constitutes a novel approach to moderation of neurodegeneration.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Huntington Disease/therapy , Semaphorins/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Brain/metabolism , Brain/pathology , Cognition Disorders/etiology , Cognition Disorders/therapy , Disease Models, Animal , Huntington Disease/complications , Immunotherapy , Mice , Mice, Transgenic , Motor Activity/drug effects , Signal Transduction/drug effects
5.
Cancer Immunol Res ; 3(6): 689-701, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25614511

ABSTRACT

Semaphorin 4D (SEMA4D, CD100) and its receptor plexin-B1 (PLXNB1) are broadly expressed in murine and human tumors, and their expression has been shown to correlate with invasive disease in several human tumors. SEMA4D normally functions to regulate the motility and differentiation of multiple cell types, including those of the immune, vascular, and nervous systems. In the setting of cancer, SEMA4D-PLXNB1 interactions have been reported to affect vascular stabilization and transactivation of ERBB2, but effects on immune-cell trafficking in the tumor microenvironment (TME) have not been investigated. We describe a novel immunomodulatory function of SEMA4D, whereby strong expression of SEMA4D at the invasive margins of actively growing tumors influences the infiltration and distribution of leukocytes in the TME. Antibody neutralization of SEMA4D disrupts this gradient of expression, enhances recruitment of activated monocytes and lymphocytes into the tumor, and shifts the balance of cells and cytokines toward a proinflammatory and antitumor milieu within the TME. This orchestrated change in the tumor architecture was associated with durable tumor rejection in murine Colon26 and ERBB2(+) mammary carcinoma models. The immunomodulatory activity of anti-SEMA4D antibody can be enhanced by combination with other immunotherapies, including immune checkpoint inhibition and chemotherapy. Strikingly, the combination of anti-SEMA4D antibody with antibody to CTLA-4 acts synergistically to promote complete tumor rejection and survival. Inhibition of SEMA4D represents a novel mechanism and therapeutic strategy to promote functional immune infiltration into the TME and inhibit tumor progression.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Neoplasms/immunology , Semaphorins/antagonists & inhibitors , Semaphorins/immunology , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Cyclophosphamide/pharmacology , Cytokines/metabolism , Disease Models, Animal , Drug Synergism , Female , Humans , Immunologic Memory , Immunomodulation/drug effects , Immunotherapy , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Tumor Burden/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
6.
Neurobiol Dis ; 73: 254-68, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25461192

ABSTRACT

Multiple sclerosis (MS) is a chronic neuroinflammatory disease characterized by immune cell infiltration of CNS, blood-brain barrier (BBB) breakdown, localized myelin destruction, and progressive neuronal degeneration. There exists a significant need to identify novel therapeutic targets and strategies that effectively and safely disrupt and even reverse disease pathophysiology. Signaling cascades initiated by semaphorin 4D (SEMA4D) induce glial activation, neuronal process collapse, inhibit migration and differentiation of oligodendrocyte precursor cells (OPCs), and disrupt endothelial tight junctions forming the BBB. To target SEMA4D, we generated a monoclonal antibody that recognizes mouse, rat, monkey and human SEMA4D with high affinity and blocks interaction between SEMA4D and its cognate receptors. In vitro, anti-SEMA4D reverses the inhibitory effects of recombinant SEMA4D on OPC survival and differentiation. In vivo, anti-SEMA4D significantly attenuates experimental autoimmune encephalomyelitis in multiple rodent models by preserving BBB integrity and axonal myelination and can be shown to promote migration of OPC to the site of lesions and improve myelin status following chemically-induced demyelination. Our study underscores SEMA4D as a key factor in CNS disease and supports the further development of antibody-based inhibition of SEMA4D as a novel therapeutic strategy for MS and other neurologic diseases with evidence of demyelination and/or compromise to the neurovascular unit.


Subject(s)
Blood-Brain Barrier/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Oligodendroglia/metabolism , Semaphorins/metabolism , Animals , Antibodies, Monoclonal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Semaphorins/antagonists & inhibitors , Semaphorins/immunology
7.
Nat Commun ; 5: 5244, 2014 Nov 05.
Article in English | MEDLINE | ID: mdl-25370169

ABSTRACT

Mitochondrial dysfunction has been reported in both familial and sporadic Parkinson's disease (PD). However, effective therapy targeting this pathway is currently inadequate. Recent studies suggest that manipulating the processes of mitochondrial fission and fusion has considerable potential for treating human diseases. To determine the therapeutic impact of targeting these pathways on PD, we used two complementary mouse models of mitochondrial impairments as seen in PD. We show here that blocking mitochondrial fission is neuroprotective in the PTEN-induced putative kinase-1 deletion (PINK1(-/-)) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse models. Specifically, we show that inhibition of the mitochondrial fission GTPase dynamin-related protein-1 (Drp1) using gene-based and small-molecule approaches attenuates neurotoxicity and restores pre-existing striatal dopamine release deficits in these animal models. These results suggest Drp1 inhibition as a potential treatment for PD.


Subject(s)
Dopamine/metabolism , Dynamins/antagonists & inhibitors , Mitochondrial Dynamics , Parkinson Disease/therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Disease Models, Animal , Dopaminergic Neurons/metabolism , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/metabolism , Parkinson Disease/metabolism , Protein Kinases/genetics
8.
Am J Pathol ; 182(6): 2285-97, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23567638

ABSTRACT

Neuroinflammation, through production of proinflammatory molecules and activated glial cells, is implicated in Alzheimer's disease (AD) pathogenesis. One such proinflammatory mediator is tumor necrosis factor α (TNF-α), a multifunctional cytokine produced in excess and associated with amyloid ß-driven inflammation and cognitive decline. Long-term global inhibition of TNF receptor type I (TNF-RI) and TNF-RII signaling without cell or stage specificity in triple-transgenic AD mice exacerbates hallmark amyloid and neurofibrillary tangle pathology. These observations revealed that long-term pan anti-TNF-α inhibition accelerates disease, cautions against long-term use of anti-TNF-α therapeutics for AD, and urges more selective regulation of TNF signaling. We used adeno-associated virus vector-delivered siRNAs to selectively knock down neuronal TNF-R signaling. We demonstrate divergent roles for neuronal TNF-RI and TNF-RII where loss of opposing TNF-RII leads to TNF-RI-mediated exacerbation of amyloid ß and Tau pathology in aged triple-transgenic AD mice. Dampening of TNF-RII or TNF-RI+RII leads to a stage-independent increase in Iba-1-positive microglial staining, implying that neuronal TNF-RII may act nonautonomously on the microglial cell population. These results reveal that TNF-R signaling is complex, and it is unlikely that all cells and both receptors will respond positively to broad anti-TNF-α treatments at various stages of disease. In aggregate, these data further support the development of cell-, stage-, and/or receptor-specific anti-TNF-α therapeutics for AD.


Subject(s)
Alzheimer Disease/metabolism , Neurons/metabolism , Receptors, Tumor Necrosis Factor/biosynthesis , Adenoviridae/genetics , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Brain/pathology , Disease Progression , Down-Regulation/physiology , Gene Knockdown Techniques , Genetic Vectors , Male , Mice , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid/metabolism , RNA, Small Interfering/genetics , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction/physiology
9.
J Pediatr Orthop ; 32(5): 473-6, 2012.
Article in English | MEDLINE | ID: mdl-22706462

ABSTRACT

INTRODUCTION: Fractures of the distal third of the radius in children are common and most heal without incident. However, distal radial shaft malunion with apex volar angulation may lead to volar dislocation of the distal ulna with forearm supination, although it has been rarely reported. The aim of this study was to document 2 such cases and to make recommendations regarding the management of these patients. METHODS: We report the cases of 2 boys, ages 6 and 8 years, who sustained radial shaft fractures that healed with apex volar angulation and who developed intractable volar dislocation of the distal ulna in adolescence. In both cases, corrective radial osteotomy at the site of the malunion restored full stability of the distal radial-ulnar joint without the need for soft-tissue reconstruction or ulnar styloid nonunion repair. DISCUSSION: This injury pattern is rarely reported but should be considered in cases of repeated volar dislocation of the distal ulna with supination. We recommend a corrective osteotomy at the site of the malunion to restore stability to the distal radial-ulnar joint. LEVEL OF EVIDENCE: IV.


Subject(s)
Osteotomy/methods , Radius Fractures/pathology , Ulna/pathology , Adolescent , Fractures, Malunited , Humans , Joint Dislocations , Joint Instability , Male , Radius Fractures/surgery , Supination
10.
J Neuroimmune Pharmacol ; 7(1): 42-59, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21728035

ABSTRACT

Tumor Necrosis Factor-alpha (TNF-α) is a prototypic pro-inflammatory cytokine involved in the innate immune response. TNF-α ligation and downstream signaling with one of its cognate receptors, TNF-RI or TNF-RII, modulates fundamental processes in the brain including synapse formation and regulation, neurogenesis, regeneration, and general maintenance of the central nervous system (CNS). During states of chronic neuroinflammation, extensive experimental evidence implicates TNF-α as a key mediator in disease progression, gliosis, demyelination, inflammation, blood-brain-barrier deterioration, and cell death. This review explores the complex roles of TNF-α in the CNS under normal physiologic conditions and during neurodegeneration. We focus our discussion on Multiple Sclerosis, Parkinson's disease, and Alzheimer's disease, relaying the outcomes of preclinical and clinical testing of TNF-α directed therapeutic strategies, and arguing that despite the wealth of functions attributed to this central cytokine, surprisingly little is known about the cell type- and stage-specific roles of TNF-α in these debilitating disorders.


Subject(s)
Alzheimer Disease/immunology , Homeostasis/immunology , Multiple Sclerosis/immunology , Parkinson Disease/immunology , Tumor Necrosis Factor-alpha/immunology , Alzheimer Disease/metabolism , Animals , Central Nervous System/immunology , Humans , Multiple Sclerosis/metabolism , Neurogenesis/immunology , Parkinson Disease/metabolism , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/metabolism
11.
PLoS One ; 6(11): e27728, 2011.
Article in English | MEDLINE | ID: mdl-22132130

ABSTRACT

Effective regulation of transgene product in anatomically circumscribed brain tissue is dependent on the pharmacokinetics of the regulating agent, the kinetics of transcriptional activation and degradation of the transgene product. We evaluated rapamycin-regulated AAV2-GDNF expression in the rat brain (striatum). Regulated (a dual-component system: AAV2-FBZhGDNF + AAV2-TF1Nc) and constitutive (CMV-driven) expression vectors were compared. Constitutively active AAV2-GDNF directed stable GDNF expression in a dose-dependent manner and it increased for the first month, thereafter reaching a plateau that was maintained over a further 3 months. For the AAV2-regGDNF, rapamycin was administered in a 3-days on/4-days off cycle. Intraperitoneal, oral, and direct brain delivery (CED) of rapamycin were evaluated. Two cycles of rapamycin at an intraperitoneal dose of 10 mg/kg gave the highest GDNF level (2.75±0.01 ng/mg protein). Six cycles at 3 mg/kg resulted in lower GDNF values (1.36±0.3 ng/mg protein). Interestingly, CED of rapamycin into the brain at a very low dose (50 ng) induced GDNF levels comparable to a 6-week intraperitoneal rapamycin cycle. This study demonstrates the effectiveness of rapamycin regulation in the CNS. However, the kinetics of the transgene in brain tissue, the regulator dosing amount and schedule are critical parameters that influence the kinetics of accumulation and zenith of the encoded transgene product.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Sirolimus/pharmacology , Transduction, Genetic/methods , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Glial Cell Line-Derived Neurotrophic Factor/genetics , Humans , Immunohistochemistry , Kinetics , Male , Neostriatum/drug effects , Neostriatum/metabolism , Rats , Rats, Sprague-Dawley , Sirolimus/administration & dosage , Specimen Handling , Time Factors
12.
J Neurosci ; 31(49): 17788-99, 2011 Dec 07.
Article in English | MEDLINE | ID: mdl-22159095

ABSTRACT

Elevating spinal levels of neurotrophin NT-3 (NT3) while increasing expression of the NR2D subunit of the NMDA receptor using a HSV viral construct promotes formation of novel multisynaptic projections from lateral white matter (LWM) axons to motoneurons in neonates. However, this treatment is ineffective after postnatal day 10. Because chondroitinase ABC (ChABC) treatment restores plasticity in the adult CNS, we have added ChABC to this treatment and applied the combination to adult rats receiving a left lateral hemisection (Hx) at T8. All hemisected animals initially dragged the ipsilateral hindpaw and displayed abnormal gait. Rats treated with ChABC or NT3/HSV-NR2D recovered partial hindlimb locomotor function, but animals receiving combined therapy displayed the most improved body stability and interlimb coordination [Basso-Beattie-Bresnahan (BBB) locomotor scale and gait analysis]. Electrical stimulation of the left LWM at T6 did not evoke any synaptic response in ipsilateral L5 motoneurons of control hemisected animals, indicating interruption of the white matter. Only animals with the full combination treatment recovered consistent multisynaptic responses in these motoneurons indicating formation of a detour pathway around the Hx. These physiological findings were supported by the observation of increased branching of both cut and intact LWM axons into the gray matter near the injury. ChABC-treated animals displayed more sprouting than control animals and those receiving NT3/HSV-NR2D; animals receiving the combination of all three treatments showed the most sprouting. Our results indicate that therapies aimed at increasing plasticity, promoting axon growth and modulating synaptic function have synergistic effects and promote better functional recovery than if applied individually.


Subject(s)
Axons/metabolism , Chondroitin ABC Lyase/metabolism , Neuronal Plasticity/physiology , Neurotrophin 3/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Recovery of Function/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Analysis of Variance , Animals , Axons/pathology , Biotin/analogs & derivatives , Biotin/metabolism , Cells, Cultured , Chondroitin Sulfate Proteoglycans/metabolism , Dextrans/metabolism , Disease Models, Animal , Excitatory Postsynaptic Potentials , Female , Fibroblasts/metabolism , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Hyperalgesia/physiopathology , Locomotion/physiology , Rats , Rats, Sprague-Dawley , Transfection , beta-Galactosidase/metabolism
13.
Eur J Neurosci ; 34(8): 1256-67, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21995852

ABSTRACT

To encourage re-establishment of functional innervation of ipsilateral lumbar motoneurons by descending fibers after an intervening lateral thoracic (T10) hemisection (Hx), we treated adult rats with the following agents: (i) anti-Nogo-A antibodies to neutralize the growth-inhibitor Nogo-A; (ii) neurotrophin-3 (NT-3) via engineered fibroblasts to promote neuron survival and plasticity; and (iii) the NMDA-receptor 2d (NR2d) subunit via an HSV-1 amplicon vector to elevate NMDA receptor function by reversing the Mg(2+) block, thereby enhancing synaptic plasticity and promoting the effects of NT-3. Synaptic responses evoked by stimulation of the ventrolateral funiculus ipsilateral and rostral to the Hx were recorded intracellularly from ipsilateral lumbar motoneurons. In uninjured adult rats short-latency (1.7-ms) monosynaptic responses were observed. After Hx these monosynaptic responses were abolished. In the Nogo-Ab + NT-3 + NR2d group, long-latency (approximately 10 ms), probably polysynaptic, responses were recorded and these were not abolished by re-transection of the spinal cord through the Hx area. This suggests that these novel responses resulted from new connections established around the Hx. Anterograde anatomical tracing from the cervical grey matter ipsilateral to the Hx revealed increased numbers of axons re-crossing the midline below the lesion in the Nogo-Ab + NT-3 + NR2d group. The combined treatment resulted in slightly better motor function in the absence of adverse effects (e.g. pain). Together, these results suggest that the combination treatment with Nogo-Ab + NT-3 + NR2d can produce a functional 'detour' around the lesion in a laterally hemisected spinal cord. This novel combination treatment may help to improve function of the damaged spinal cord.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Myelin Proteins/immunology , Neurotrophin 3/pharmacology , Protein Subunits/pharmacology , Receptors, N-Methyl-D-Aspartate/therapeutic use , Spinal Cord Injuries/pathology , Spinal Cord/drug effects , Animals , Behavior, Animal/physiology , Female , Humans , Motor Activity/drug effects , Motor Activity/physiology , Motor Neurons/cytology , Motor Neurons/drug effects , Motor Neurons/physiology , Neurotrophin 3/therapeutic use , Nogo Proteins , Protein Subunits/therapeutic use , Psychomotor Performance , Rats , Rats, Sprague-Dawley , Spinal Cord/pathology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/physiopathology
14.
Vaccine ; 29(40): 7080-9, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21801777

ABSTRACT

The efficient induction of virus-specific mucosal antibodies is an important unmet objective in Human Immunodeficiency Virus Type-1 (HIV-1) vaccine research. One promising approach is sublingual (SL) immunization. We examined the effectiveness of SL delivery of two different viral vectors: (i) a recombinant adenovirus (rAd5), and (ii) a Herpes Simplex Virus Type-1 amplicon vector (HSV-1). Initial in vitro videomicroscopy experiments showed that rAd5 particles were trapped in saliva (i.e., that Ad5 was mucoadhesive) - unlike HSV-1 virions, which migrated freely in both saliva and water. In vivo imaging studies in mice revealed that only the rAd5 vector efficiently transduced the SL epithelium. Consistent with this, SL delivery of an rAd5 encoding HIV-1 envelope glycoprotein (Env) resulted in robust antigen-specific antibody responses in plasma and in vaginal washes, whereas SL delivery of a HSV-1 amplicon vector encoding HIV-1 Env failed to elicit Env-specific antibodies. In contrast, both vectors elicited equivalent humoral responses following intramuscular (IM) delivery. Finally, SL delivery of the rAd5:Env vector resulted in elevated levels of Env-specific serum IgA, and vaginal IgA and IgG, when compared to IM delivery of the same vector. These results findings shed light on vector properties (mucoadhesion, penetration of the sublingual barrier) which may be important for the induction of potent humoral immune responses following sublingual vector administration. Our data also show that SL delivery of an Env-encoding rAd5 vector can elicit a potent antigen-specific mucosal antibody response in the absence of adjuvant. Overall, these findings support the further exploration of the SL delivery route for HIV-1 vaccine delivery.


Subject(s)
AIDS Vaccines/administration & dosage , AIDS Vaccines/immunology , HIV-1/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/genetics , AIDS Vaccines/pharmacokinetics , Adenoviridae/genetics , Adenoviridae/immunology , Administration, Sublingual , Animals , Antibodies, Neutralizing/immunology , Cell Line , Female , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/immunology , HEK293 Cells , HIV Antibodies/blood , HIV Antibodies/immunology , HIV-1/genetics , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Humans , Immunity, Humoral/immunology , Immunity, Mucosal/genetics , Immunity, Mucosal/immunology , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Saliva/immunology , Virion/immunology , env Gene Products, Human Immunodeficiency Virus/genetics
15.
Am J Pathol ; 179(4): 2053-70, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21835156

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by severe memory loss and cognitive impairment. Neuroinflammation, including the extensive production of pro-inflammatory molecules and the activation of microglia, has been implicated in the disease process. Tumor necrosis factor (TNF)-α, a prototypic pro-inflammatory cytokine, is elevated in AD, is neurotoxic, and colocalizes with amyloid plaques in AD animal models and human brains. We previously demonstrated that the expression of TNF-α is increased in AD mice at ages preceding the development of hallmark amyloid and tau pathological features and that long-term expression of this cytokine in these mice leads to marked neuronal death. Such observations suggest that TNF-α signaling promotes AD pathogenesis and that therapeutics suppressing this cytokine's activity may be beneficial. To dissect TNF-α receptor signaling requirements in AD, we generated triple-transgenic AD mice (3xTg-AD) lacking both TNF-α receptor 1 (TNF-RI) and 2 (TNF-RII), 3xTg-ADxTNF-RI/RII knock out, the cognate receptors of TNF-α. These mice exhibit enhanced amyloid and tau-related pathological features by the age of 15 months, in stark contrast to age-matched 3xTg-AD counterparts. Moreover, 3xTg-ADxTNF-RI/RII knock out-derived primary microglia reveal reduced amyloid-ß phagocytic marker expression and phagocytosis activity, indicating that intact TNF-α receptor signaling is critical for microglial-mediated uptake of extracellular amyloid-ß peptide pools. Overall, our results demonstrate that globally ablated TNF receptor signaling exacerbates pathogenesis and argues against long-term use of pan-anti-TNF-α inhibitors for the treatment of AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type I/deficiency , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Aging/pathology , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Amyloid/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , CA3 Region, Hippocampal/pathology , CA3 Region, Hippocampal/physiopathology , Calcium-Binding Proteins/metabolism , Crosses, Genetic , Female , Humans , Lipopolysaccharide Receptors/metabolism , Long-Term Potentiation , Male , Mice , Mice, Knockout , Microfilament Proteins/metabolism , Microglia/metabolism , Microglia/pathology , Phagocytosis , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Synapses/metabolism , Transgenes/genetics , Tumor Necrosis Factor-alpha/metabolism , tau Proteins/genetics
16.
J Alzheimers Dis ; 27(2): 361-76, 2011.
Article in English | MEDLINE | ID: mdl-21860086

ABSTRACT

Transgenic (Tg) mouse models of Alzheimer's disease (AD) have been genetically altered with human familial AD genes driven by powerful promoters. However, a Tg model must accurately mirror the pathogenesis of the human disease, not merely the signature amyloid and/or tau pathology, as such hallmarks can arise via multiple convergent or even by pathogenic mechanisms unrelated to human sporadic AD. The 3 × Tg-AD mouse simultaneously expresses 3 rare familial mutant genes that in humans independently produce devastating amyloid-ß protein precursor (AßPP), presenilin-1, and frontotemporal dementias; hence, technically speaking, these mice are not a model of sporadic AD, but are informative in assessing co-evolving amyloid and tau pathologies. While end-stage amyloid and tau pathologies in 3 × Tg-AD mice are similar to those observed in sporadic AD, the pathophysiological mechanisms leading to these lesions are quite different. Comprehensive biochemical and morphological characterizations are important to gauge the predictive value of Tg mice. Investigation of AßPP, amyloid-ß (Aß), and tau in the 3 × Tg-AD model demonstrates AD-like pathology with some key differences compared to human sporadic AD. The biochemical dissection of AßPP reveals different cleavage patterns of the C-terminus of AßPP when compared to human AD, suggesting divergent pathogenic mechanisms. Human tau is concomitantly expressed with AßPP/Aß from an early age while abundant extracellular amyloid plaques and paired helical filaments are manifested from 18 months on. Understanding the strengths and limitations of Tg mouse AD models through rigorous biochemical, pathological, and functional analyses will facilitate the derivation of models that better approximate human sporadic AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/biosynthesis , Disease Models, Animal , Presenilin-1/genetics , tau Proteins/genetics , Aged, 80 and over , Alzheimer Disease/genetics , Animals , Female , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Presenilin-1/biosynthesis , tau Proteins/biosynthesis
17.
Curr Gene Ther ; 11(5): 332-40, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21711226

ABSTRACT

The pursuits of sustainable treatments for diseases and disorders that afflict the central nervous system (CNS) have proven challenging for the field of viral vector-based gene therapy. However, recent advances in viral vector technology coupled with efficient delivery methods have opened up new avenues that show promise at the preclinical testing stage. The development of the Herpes Simplex Virus/Sleeping Beauty (HSV/SB) hybrid vector represents such an advance for devising treatments targeting the CNS with its potential for stably integrating large transgenomic segments of DNA within the genomes of transduced cells. In utero administration of this hybrid vector into the embryonic mouse brain has revealed the capacity for widespread transgene dissemination due to the targeting of a neuronal precursor cell population. This unique feature has provided the means to stably express a transgene throughout the brain for prolonged periods, which is a prerequisite for the treatment of progressive CNS disorders. In this review we provide a comprehensive breakdown of the characteristics of the HSV/SB vector system and how it can be efficiently employed in the derivation of CNS-targeted gene therapeutic strategies.


Subject(s)
Central Nervous System Diseases/therapy , Gene Targeting/methods , Genetic Therapy/methods , Genetic Vectors/genetics , Neural Stem Cells , Simplexvirus/genetics , Animals , Blindness/therapy , Brain/embryology , Brain/growth & development , Genome, Human , Humans , Mice , Transgenes/genetics , Transposases/genetics
18.
Hum Mol Genet ; 20(R1): R28-41, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21429918

ABSTRACT

Genetic therapy is undergoing a renaissance with expansion of viral and synthetic vectors, use of oligonucleotides (RNA and DNA) and sequence-targeted regulatory molecules, as well as genetically modified cells, including induced pluripotent stem cells from the patients themselves. Several clinical trials for neurologic syndromes appear quite promising. This review covers genetic strategies to ameliorate neurologic syndromes of different etiologies, including lysosomal storage diseases, Alzheimer's disease and other amyloidopathies, Parkinson's disease, spinal muscular atrophy, amyotrophic lateral sclerosis and brain tumors. This field has been propelled by genetic technologies, including identifying disease genes and disruptive mutations, design of genomic interacting elements to regulate transcription and splicing of specific precursor mRNAs and use of novel non-coding regulatory RNAs. These versatile new tools for manipulation of genetic elements provide the ability to tailor the mode of genetic intervention to specific aspects of a disease state.


Subject(s)
Genetic Therapy/methods , Nervous System Diseases/therapy , Neurodegenerative Diseases/therapy , Alzheimer Disease/therapy , Genetic Vectors/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Lysosomal Storage Diseases/therapy , Parkinson Disease/therapy , RNA Splicing
19.
PLoS One ; 6(1): e16222, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21283759

ABSTRACT

T lymphocytes of the CD8+ class are critical in delivering cytotoxic function and in controlling viral and intracellular infections. These cells are "helped" by T lymphocytes of the CD4+ class, which facilitate their activation, clonal expansion, full differentiation and the persistence of memory. In this study we investigated the impact of CD4+ T cells on the location of CD8+ T cells, using antibody-mediated CD4+ T cell depletion and imaging the antigen-driven redistribution of bioluminescent CD8+ T cells in living mice. We documented that CD4+ T cells influence the biodistribution of CD8+ T cells, favoring their localization to abdominal lymph nodes. Flow cytometric analysis revealed that this was associated with an increase in the expression of specific integrins. The presence of CD4+ T cells at the time of initial CD8+ T cell activation also influences their biodistribution in the memory phase. Based on these results, we propose the model that one of the functions of CD4+ T cell "help" is to program the homing potential of CD8+ T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Cell Movement/immunology , Animals , Antigens/pharmacology , CD8-Positive T-Lymphocytes/cytology , Immunologic Memory , Integrins/analysis , Luminescent Measurements , Lymph Nodes/immunology , Mice , Tissue Distribution
20.
Glia ; 59(4): 627-40, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21294162

ABSTRACT

White matter pathology has been documented in the brains of familial Alzheimer's disease (FAD)-afflicted individuals during presymptomatic and preclinical stages of AD. How these defects in myelination integrity arise and what roles they may play in AD pathophysiology have yet to be fully elucidated. We previously demonstrated that triple-transgenic AD (3xTg-AD) mice, which harbor the human amyloid precursor Swedish mutation, presenilin-1 M146V (PS1(M146V) ) knock-in mutation, and tau(P301L) mutation, exhibit myelin abnormalities analogous to FAD patients and that Aß(1-42) contributes to these white matter deficits. Herein, we demonstrate that the PS1(M146V) mutation predisposes mouse oligodendrocyte precursor (mOP) cells to Aß(1-42) -induced alterations in cell differentiation in vitro. Furthermore, PS1(M146V) expression compromised mOP cell function and MBP protein distribution, a process that is further aggravated with exposure to Aß(1-42) . We found that the myelination defect and MBP subcellular mislocalization triggered by PS1(M146V) and Aß(1-42) can be effectively prevented by treatment with the GSK-3ß inhibitor, TWS119, thereby implicating GSK-3ß kinase activity in this pathogenic cascade. Overall, this work provides further mechanistic insights into PS1(M146V) and Aß(1-42) -driven oligodendrocyte dysfunction andmyelin damage during early presymptomatic stages of AD, and provides a new target in oligodendrocytes for developing therapies designed to avert AD-related white matter pathology.


Subject(s)
Amyloid beta-Peptides/metabolism , Cell Differentiation/physiology , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Presenilin-1/genetics , Amyloid beta-Peptides/pharmacology , Analysis of Variance , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Line , Flow Cytometry , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Immunohistochemistry , Mice , Mice, Transgenic , Mutation , Myelin Sheath/drug effects , Myelin Sheath/genetics , Myelin Sheath/pathology , Oligodendroglia/drug effects , Oligodendroglia/pathology , Presenilin-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...