Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
J Inorg Biochem ; 203: 110860, 2020 02.
Article in English | MEDLINE | ID: mdl-31698325

ABSTRACT

The first successful attempt to obtain purified aluminum metal was accomplished by the Danish physicist and chemist Hans Christian Orsted in 1824, however it was not until about ~140 years later that aluminum's capacity for neurological disruption and neurotoxicity was convincingly established. The earliest evidence of the possible involvement of this biosphere-rich metallotoxin in Alzheimer's disease (AD) originated in the early-to-mid-1960's from animal and human research investigations that arose almost simultaneously from independent laboratories in the United States and Canada. This short communication pays tribute to the pioneering research work on aluminum in susceptible species, in AD animal models and in AD patients by the early investigators Drs. Robert D. Terry, Igor Klatzo and Henryk M. Wisniewski with special acknowledgement to the late Dr. Donald RC McLachlan, and their contemporary physician-scientist colleagues and collaborators. Together these researchers established the groundwork and foundation towards our understanding of the potential contribution of aluminum to progressive, age-related and lethal neurodegenerative diseases of the human central nervous system.


Subject(s)
Aluminum/toxicity , Neurosciences/history , Neurotoxicity Syndromes/etiology , Alzheimer Disease/etiology , Amyloid/drug effects , Animals , Brain/pathology , History, 20th Century , History, 21st Century , Humans , Neurofibrillary Tangles/drug effects , Plaque, Amyloid/etiology , United States
2.
J Inorg Biochem ; 203: 110886, 2020 02.
Article in English | MEDLINE | ID: mdl-31707334

ABSTRACT

Gram-negative bacteria of the human gastrointestinal (GI) tract microbiome: (i) are capable of generating a broad-spectrum of highly neurotoxic, pro-inflammatory and potentially pathogenic molecules; and (ii) these include a highly immunogenic class of amphipathic surface glycolipids known as lipopolysaccharide (LPS). Bacteroides fragilis (B. fragilis), a commensal, Gram negative, non-motile, non-spore forming obligatory anaerobic bacillus, and one of the most abundant bacteria found in the human GI tract, produces a particularly pro-inflammatory and neurotoxic LPS (BF-LPS). BF-LPS: (i) is known to be secreted from the B. fragilis outer membrane into the external-medium; (ii) can damage biophysiological barriers via cleavage of zonula adherens cell-cell adhesion proteins, thereby disrupting both the GI-tract barrier and the blood-brain barrier (BBB); (iii) is able to transit GI-tract barriers into the systemic circulation and cross the BBB into the human CNS; and (iv) accumulates within CNS neurons in neurodegenerative disorders such as Alzheimer's disease (AD). This short communication provides evidence that the incubation of B. fragilis with aluminum sulfate [Al2(SO4)3] is a potent inducer of BF-LPS. The results suggest for the first time that the pro-inflammatory properties of aluminum may not only be propagated by aluminum itself, but by a stimulation in the production of microbiome-derived BF-LPS and other pro-inflammatory pathogenic microbial products normally secreted from human GI-tract-resident microorganisms.


Subject(s)
Alum Compounds/pharmacology , Bacteroides fragilis/drug effects , Lipopolysaccharides/metabolism , Bacteroides fragilis/metabolism
3.
Biochemistry (Mosc) ; 83(9): 1018-1029, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30472940

ABSTRACT

Both plants and animals have adopted a common strategy of using ~18-25-nucleotide small non-coding RNAs (sncRNAs), known as microRNAs (miRNAs), to transmit DNA-based epigenetic information. miRNAs (i) shape the total transcriptional output of individual cells; (ii) regulate and fine-tune gene expression profiles of cell clusters, and (iii) modulate cell phenotype in response to environmental stimuli and stressors. These miRNAs, the smallest known carriers of gene-encoded post-transcriptional regulatory information, not only regulate cellular function in healthy cells but also act as important mediators in the development of plant and animal diseases. Plants possess their own specific miRNAs; at least 32 plant species have been found to carry infectious sncRNAs called viroids, whose mechanisms of generation and functions are strikingly similar to those of miRNAs. This review highlights recent remarkable and sometimes controversial findings in miRNA signaling in plants and animals. Special attention is given to the intriguing possibility that dietary miRNAs and/or sncRNAs can function as mobile epigenetic and/or evolutionary linkers between different species and contribute to both intra- and interkingdom signaling. Wherever possible, emphasis has been placed on the relevance of these miRNAs to the development of human neurodegenerative diseases, such as Alzheimer's disease. Based on the current available data, we suggest that such xeno-miRNAs may (i) contribute to the beneficial properties of medicinal plants, (ii) contribute to the negative properties of disease-causing or poisonous plants, and (iii) provide cross-species communication between kingdoms of living organisms involving multiple epigenetic and/or potentially pathogenic mechanisms associated with the onset and pathogenesis of various diseases.


Subject(s)
MicroRNAs/metabolism , Neurodegenerative Diseases/pathology , Plants/genetics , Viroids/physiology , Animals , Central Nervous System/metabolism , Gene Expression Regulation , Humans , Neurodegenerative Diseases/genetics , Plants/metabolism
4.
Article in English | MEDLINE | ID: mdl-29051843

ABSTRACT

RNA sequencing, DNA microfluidic array, LED-Northern, Western immunoassay and bioinformatics analysis have uncovered a small family of up-regulated human brain enriched microRNAs (miRNAs) and down-regulated messenger RNAs (mRNAs) in short post-mortem interval (PMI) sporadic Alzheimer's disease (AD) brain. At the mRNA level, a large majority of the expression of human brain genes found to be down-regulated in sporadic AD appears to be a consequence of an up-regulation of a specific group of NF-kB-inducible microRNAs (miRNAs). This group of up-regulated miRNAs - including miRNA-34a and miRNA-146a - has strong, energetically favorable, complimentary RNA sequences in the 3' untranslated regions (3'-UTR) of their target mRNAs which ultimately drive the down-regulation in the expression of certain essential brain genes. Interestingly, just 2 significantly up-regulated miRNAs - miRNA-34a and miRNA-146a - appear to down-regulate mRNA targets involved in synaptogenesis (SHANK3), phagocytosis deficits and tau pathology (TREM2), inflammation (CFH; complement factor H) and amyloidogenesis (TSPAN12), all of which are distinguishing pathological features characteristic of middle-to-late stage AD neuropathology. This paper reports the novel finding of parallel miRNA-34a and miRNA-146a up-regulation in sporadic AD hippocampal CA1 RNA pools and proposes an altered miRNA-mRNA coupled signaling network in AD, much of which is supported by current experimental findings in the recent literature.

5.
Biochemistry (Mosc) ; 82(2): 122-139, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28320296

ABSTRACT

Certain cellular proteins normally soluble in the living organism under certain conditions form aggregates with a specific cross-ß sheet structure called amyloid. These intra- or extracellular insoluble aggregates (fibers or plaques) are hallmarks of many neurodegenerative pathologies including Alzheimer's disease (AD), Huntington's disease, Parkinson's disease, prion disease, and other progressive neurological diseases that develop in the aging human central nervous system. Amyloid diseases (amyloidoses) are widespread in the elderly human population, a rapidly expanding demographic in many global populations. Increasing age is the most significant risk factor for neurodegenerative diseases associated with amyloid plaques. To date, nearly three dozen different misfolded proteins targeting brain and other organs have been identified in amyloid diseases and AD, the most prevalent neurodegenerative amyloid disease affecting over 15 million people worldwide. Here we (i) highlight the latest data on mechanisms of amyloid formation and further discuss a hypothesis on the amyloid cascade as a primary mechanism of AD pathogenesis and (ii) review the evolutionary aspects of amyloidosis, which allow new insight on human-specific mechanisms of dementia development.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Amyloid/metabolism , Age Factors , Aging/pathology , Alzheimer Disease/epidemiology , Alzheimer Disease/pathology , Animals , Humans , Risk Factors
6.
Article in English | MEDLINE | ID: mdl-29354323

ABSTRACT

A number of experimental investigations utilizing different murine species have previously reported: (i) that standard mouse-diets supplemented with physiologically realistic amounts of neurotoxic metal salts substantially induce pro-inflammatory signaling in a number of murine tissues; (ii) that these diet-stimulated changes may contribute to a systemic inflammation (SI), a potential precursor to neurodegenerative events in both the central and the peripheral nervous system (CNS, PNS); and (iii) that these events may ultimately contribute to a chronic and progressive inflammatory neurodegeneration, such as that which is observed in Alzheimer's disease (AD) brain. In these experiments we assayed for markers of SI in the blood serum of C57BL/6J mice after 0, 1, 3 and 5 months of exposure to a standard mouse diet that included aluminum-sulfate in the food and drinking water, compared to age-matched controls receiving magnesium-sulfate or no additions. The data indicate that the SI markers that include the pro-inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor alpha (TNFα), the acute phase reactive protein C-reactive protein (CRP) production and a triad of pro-inflammatory microRNAs (miRNA-9, miRNA-125b and miRNA-146a) all increase in the serum after aluminum-sulfate exposure. For the first time these results suggest that ad libitum exposure to aluminum-sulfate at physiologically realistic concentrations, as would be found in the human diet over the long term, may predispose to SI and the potential development of chronic, progressive, inflammatory neurodegeneration with downstream pathogenic consequences.

7.
Morphologie ; 100(329): 56-64, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26969391

ABSTRACT

The genomes of eukaryotes orchestrate their expression to ensure an effective, homeostatic and functional gene signaling program, and this includes fundamentally altered patterns of transcription during aging, development, differentiation and disease. These actions constitute an extremely complex and intricate process as genetic operations such as transcription involve the very rapid translocation and polymerization of ribonucleotides using RNA polymerases, accessory transcription protein complexes and other interrelated chromatin proteins and genetic factors. As both free ribonucleotides and polymerized single-stranded RNA chains, ribonucleotides are highly charged with phosphate, and this genetic system is extremely vulnerable to disruption by a large number of electrostatic forces, and primarily by cationic metals such as aluminum. Aluminum has been shown by independent researchers to be particularly genotoxic to the genetic apparatus, and it has become reasonably clear that aluminum disturbs genetic signaling programs in the CNS that bear a surprising resemblance to those observed in Alzheimer's disease (AD) brain. This paper will focus on a discussion of two molecular-genetic aspects of aluminum genotoxicity: (1) the observation that micro-RNA (miRNA)-mediated global gene expression patterns in aluminum-treated transgenic animal models of AD (Tg-AD) strongly resemble those found in AD; and (2) the concept of "human biochemical individuality" and the hypothesis that individuals with certain gene expression patterns may be especially sensitive and perhaps predisposed to aluminum genotoxicity.


Subject(s)
Aluminum Compounds/toxicity , Alzheimer Disease/genetics , Central Nervous System/drug effects , DNA Damage/drug effects , MicroRNAs/metabolism , Mutagens/toxicity , Aging/genetics , Alzheimer Disease/chemically induced , Animals , Disease Models, Animal , Gene Expression Profiling , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Mice , Mice, Transgenic
8.
J Inorg Biochem ; 152: 206-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26213226

ABSTRACT

At least 57 murine transgenic models for Alzheimer's disease (Tg-AD) have been developed to overexpress the 42 amino acid amyloid-beta (Aß42) peptide in the central nervous system (CNS). These 'humanized murine Tg-AD models' have greatly expanded our understanding of the contribution of Aß42 peptide-mediated pro-inflammatory neuropathology to the AD process. A number of independent laboratories using different amyloid-overexpressing Tg-AD models have shown that supplementation of murine Tg-AD diets and/or drinking water with aluminum significantly enhances Aß42 peptide-mediated inflammatory pathology and AD-type cognitive change compared to animals receiving control diets. In humans AD-type pathology appears to originate in the limbic system and progressively spreads into primary processing and sensory regions such as the retina. In these studies, for the first time, we assess the propagation of Aß42 and inflammatory signals into the retina of 5xFAD Tg-AD amyloid-overexpressing mice whose diets were supplemented with aluminum. The two most interesting findings were (1) that similar to other Tg-AD models, there was a significantly accelerated development of Aß42 and inflammatory pathology in 5xFAD Tg-AD mice fed aluminum; and (2) in aluminum-supplemented animals, markers for inflammatory pathology appeared in both the brain and the retina as evidenced by an evolving presence of Aß42 peptides, and accompanied by inflammatory markers - cyclooxygenase-2 (COX-2) and C-reactive protein (CRP). The results indicate that in the 5xFAD Tg-AD model aluminum not only enhances an Aß42-mediated inflammatory degeneration of the brain but also appears to induce AD-type pathology in an anatomically-linked primary sensory area that involves vision.


Subject(s)
Aluminum Compounds/toxicity , Alzheimer Disease/pathology , Protein Aggregation, Pathological/chemically induced , Retina/drug effects , Aluminum Compounds/adverse effects , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/pathology , C-Reactive Protein/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , Mice , Mice, Inbred C57BL , Peptide Fragments/metabolism , Retina/pathology
9.
J Alzheimers Dis Parkinsonism ; 5(1): 177, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25977840

ABSTRACT

Since the inception of the human microbiome project (HMP) by the US National Institutes of Health (NIH) in 2007 there has been a keen resurgence in our recognition of the human microbiome and its contribution to development, immunity, neurophysiology, metabolic and nutritive support to central nervous system (CNS) health and disease. What is not generally appreciated is that (i) the ~1014 microbial cells that comprise the human microbiome outnumber human host cells by approximately one hundred-to-one; (ii) together the microbial genes of the microbiome outnumber human host genes by about one hundred-and-fifty to one; (iii) collectively these microbes constitute the largest 'diffuse organ system' in the human body, more metabolically active than the liver; strongly influencing host nutritive-, innate-immune, neuroinflammatory-, neuromodulatory- and neurotransmission-functions; and (iv) that these microbes actively secrete highly complex, immunogenic mixtures of lipopolysaccharide (LPS) and amyloid from their outer membranes into their immediate environment. While secreted LPS and amyloids are generally quite soluble as monomers over time they form into highly insoluble fibrous protein aggregates that are implicated in the progressive degenerative neuropathology of several common, age-related disorders of the human CNS including Alzheimer's disease (AD). This general commentary-perspective paper will highlight some recent findings on microbial-derived secreted LPS and amyloids and the potential contribution of these neurotoxic and proinflammatory microbial exudates to age-related inflammatory amyloidogenesis and neurodegeneration, with specific reference to AD wherever possible.

10.
Med Hypotheses ; 79(5): 622-6, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22959996

ABSTRACT

Glial neoplasms account for nearly 50% of all adult primary brain tumors. They originate from glial cells in the brain and/or spinal cord and include low-grade diffuse astrocytomas, anaplastic-astrocytomas, and glioblastomas. Of all brain tumors, glioblastoma multiforme (GBM) is the most aggressive and is characterized by rapid glial cell growth, resistance to radio- and chemo- therapies, and relentless infiltration and spreading throughout the central nervous system (CNS). In glioblastomas, primary tumor growth and CNS invasion are associated with the activation of complex structural molecular and metabolic changes within the tumor tissue, which profoundly affect the surrounding neuronal networks and may in part explain induction of epilepsy. In fact, epileptic seizures are very common among patients with glial tumors, reaching nearly 50% in glioblastoma patients and almost 90% in low-grade astrocytomas. The overall hypothesis presented here discusses the possibility that the aberrant tumor cell metabolism may act directly on neuronal network, and this leads to seizure susceptibility. Further invasion and growth of the malignant glial cells exacerbate this initial pathologic state which promotes recurrent seizures (epileptogenesis).


Subject(s)
Brain Neoplasms/complications , Glioma/complications , Seizures/complications , Humans , Models, Theoretical
11.
J Toxicol Environ Health A ; 74(22-24): 1460-8, 2011.
Article in English | MEDLINE | ID: mdl-22043907

ABSTRACT

A mouse- and human-brain-abundant, nuclear factor (NF)-кB-regulated, micro RNA-146a (miRNA-146a) is an important modulator of the innate immune response and inflammatory signaling in specific immunological and brain cell types. Levels of miRNA-146a are induced in human brain cells challenged with at least five different species of single- or double-stranded DNA or RNA neurotrophic viruses, suggesting a broad role for miRNA-146a in the brain's innate immune response and antiviral immunity. Upregulated miRNA-146a is also observed in pro-inflammatory cytokine-, Aß42 peptide- and neurotoxic metal-induced, oxidatively stressed human neuronal-glial primary cell cocultures, in murine scrapie and in Alzheimer's disease (AD) brain. In AD, miRNA-146a levels are found to progressively increase with disease severity and co-localize to brain regions enriched in inflammatory neuropathology. This study provides evidence of upregulation of miRNA-146a in extremely rare (incidence 1-10 per 100 million) human prion-based neurodegenerative disorders, including sporadic Creutzfeldt-Jakob disease (sCJD) and Gerstmann-Straussler-Scheinker syndrome (GSS). The findings suggest that an upregulated miRNA-146a may be integral to innate immune or inflammatory brain cell responses in prion-mediated infections and to progressive and irreversible neurodegeneration of both the murine and human brain.


Subject(s)
Amyloid beta-Peptides/therapeutic use , Creutzfeldt-Jakob Syndrome/drug therapy , Gerstmann-Straussler-Scheinker Disease/drug therapy , MicroRNAs/drug effects , Neurogenic Inflammation/drug therapy , Peptide Fragments/therapeutic use , Up-Regulation/drug effects , Amyloid beta-Peptides/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cells, Cultured , Creutzfeldt-Jakob Syndrome/metabolism , Creutzfeldt-Jakob Syndrome/pathology , Gerstmann-Straussler-Scheinker Disease/metabolism , Gerstmann-Straussler-Scheinker Disease/pathology , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neurogenic Inflammation/metabolism , Neurogenic Inflammation/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Peptide Fragments/pharmacology
12.
Neurosci Lett ; 487(1): 94-8, 2011 Jan 03.
Article in English | MEDLINE | ID: mdl-20934487

ABSTRACT

A mouse and human brain-enriched micro-RNA-146a (miRNA-146a) is known to be important in modulating the innate immune response and inflammatory signaling in certain immunological and brain cell types. In this study we examined miRNA-146a levels in early-, moderate- and late-stage Alzheimer's disease (AD) neocortex and hippocampus, in several human primary brain and retinal cell lines, and in 5 different transgenic mouse models of AD including Tg2576, TgCRND8, PSAPP, 3xTg-AD and 5xFAD. Inducible expression of miRNA-146a was found to be significantly up-regulated in a primary co-culture of human neuronal-glial (HNG) cells stressed using interleukin1-beta (IL-1ß), and this up-regulation was quenched using specific NF-кB inhibitors including curcumin. Expression of miRNA-146a correlated with senile plaque density and synaptic pathology in Tg2576 and in 5xFAD transgenic mouse models used in the study of this common neurodegenerative disorder.


Subject(s)
Alzheimer Disease/metabolism , Hippocampus/metabolism , MicroRNAs/metabolism , Neocortex/metabolism , Up-Regulation/genetics , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Cell Line, Transformed , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Humans , Mice , Mice, Transgenic , MicroRNAs/genetics , Mutation , Presenilin-1/genetics , Transfection/methods , Up-Regulation/drug effects , tau Proteins/genetics
13.
Neurosci Lett ; 476(1): 18-22, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20347935

ABSTRACT

Micro RNAs (miRNAs) are post-transcriptional modulators of gene expression that regulate the stability and translation of their target messenger RNAs (mRNAs). Here we report that the levels of a human brain-enriched miRNA-125b are up-regulated in interleukin-6 (IL-6)-stressed normal human astrocytes (NHA), a treatment known to induce astrogliosis. In vitro, anti-miRNA-125b added exogenously to IL-6-stressed NHA cultures attenuated both glial cell proliferation and increased the expression of the cyclin-dependent kinase inhibitor 2A (CDKN2A), a miRNA-125b target and negative regulator of cell growth. A strong positive correlation between miRNA-125b abundance and the glial cell markers glial fibrillary acidic protein (GFAP) and vimentin, and CDKN2A down-regulation was noted in advanced Alzheimer's disease (AD) and in Down's syndrome (DS) brain, chronic neurological disorders associated with astrogliosis. The results suggest that miRNA-125b up-regulation contributes to astrogliosis and to defects in the cell cycle that are characteristic of degenerating brain tissues.


Subject(s)
Astrocytes/metabolism , MicroRNAs/biosynthesis , Actins/metabolism , Astrocytes/cytology , Astrocytes/drug effects , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans , Interleukin-6/pharmacology , MicroRNAs/pharmacology , Up-Regulation , Vimentin/metabolism
14.
J Neurooncol ; 98(3): 297-304, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19941032

ABSTRACT

High density micro-RNA (miRNA) arrays, fluorescent-reporter miRNA assay and Northern miRNA dot-blot analysis show that a brain-enriched miRNA-128 is significantly down-regulated in glioblastoma multiforme (GBM) and in GBM cell lines when compared to age-matched controls. The down-regulation of miRNA-128 was found to inversely correlate with WHO tumor grade. Three bioinformatics-verified miRNA-128 targets, angiopoietin-related growth factor protein 5 (ARP5; ANGPTL6), a transcription suppressor that promotes stem cell renewal and inhibits the expression of known tumor suppressor genes involved in senescence and differentiation, Bmi-1, and a transcription factor critical for the control of cell-cycle progression, E2F-3a, were found to be up-regulated. Addition of exogenous miRNA-128 to CRL-1690 and CRL-2610 GBM cell lines (a) restored 'homeostatic' ARP5 (ANGPTL6), Bmi-1 and E2F-3a expression, and (b) significantly decreased the proliferation of CRL-1690 and CRL-2610 cell lines. Our data suggests that down-regulation of miRNA-128 may contribute to glioma and GBM, in part, by coordinately up-regulating ARP5 (ANGPTL6), Bmi-1 and E2F-3a, resulting in the proliferation of undifferentiated GBM cells.


Subject(s)
Angiopoietins/metabolism , Brain Neoplasms/pathology , Brain/pathology , Cell Proliferation , Down-Regulation/physiology , E2F3 Transcription Factor/metabolism , Glioblastoma/pathology , MicroRNAs/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Adult , Angiopoietin-Like Protein 6 , Angiopoietin-like Proteins , Angiopoietins/genetics , Cell Proliferation/drug effects , Computational Biology/methods , Down-Regulation/drug effects , E2F3 Transcription Factor/genetics , Female , Humans , Male , MicroRNAs/genetics , MicroRNAs/pharmacology , Middle Aged , Nuclear Proteins/genetics , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Transfection/methods
15.
J Neurooncol ; 91(1): 27-32, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18759060

ABSTRACT

Glioblastoma multiforme (GBM) represents a class of malignant gliomas which rapidly proliferate, invade and destroy surrounding brain tissues. This study examined micro-RNA (miRNA) speciation and miRNA effects on gene expression in six ATCC glioma and GBM cell lines and in 14 glioma and GBM samples obtained from human brain biopsy. We observed selective up-regulation of miRNA-221 and down-regulation of a miRNA-221 messenger RNA target encoding the survivin-1 homolog BIRC1, a neuronal inhibitor of apoptosis protein (NIAP) and marker for neurodegeneration. The expression of BIRC5 (survivin-1) and caspase-3 were found to be significantly up-regulated, particularly in stage IV GBM. These studies suggest that the abundance and speciation of the BIRC family of neural cell fate regulators are differentially regulated in glioma and GBM, and may contribute to progressive changes in apoptotic signaling and altered neural cell cycling functions.


Subject(s)
Caspase 3/metabolism , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/metabolism , MicroRNAs/metabolism , Neuronal Apoptosis-Inhibitory Protein/metabolism , Adult , Analysis of Variance , Biopsy/methods , Female , Glioblastoma/genetics , Glioma/genetics , Glioma/metabolism , Humans , Inhibitor of Apoptosis Proteins , Male , MicroRNAs/genetics , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Middle Aged , Neuronal Apoptosis-Inhibitory Protein/genetics , RNA, Messenger/metabolism , Survivin
16.
Biochem Soc Trans ; 34(Pt 6): 1277-82, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17073801

ABSTRACT

Significant advancements in our understanding of cell-survival signalling in AD (Alzheimer's disease) stem from recent investigations into the metabolism, trafficking and fate of the essential omega-3 fatty acid DHA (docosahexaenoic acid) (C(22:6), n=3). Brain synaptic terminals and neuronal plasma membranes are highly enriched in DHA, and deficiencies in this polyunsaturated fatty acid are characteristic of AD-affected brain. Oxidative stress, targeting phospholipids containing DHA, and age-related DHA depletion are associated with the progressive erosion of normal cognitive function in AD. Current studies support the idea that DHA itself and novel DHA-derived neural synapse- and membrane-derived lipid messengers have considerable potential to modulate cell survival signalling in stressed cultured neural cell models in vitro and in mammalian models of learning, memory and AD in vivo. Key players in this intrinsic rescue system include the alpha-secretase-processed neurotrophin sAPPalpha [soluble APPalpha (amyloid precursor protein alpha)] peptide, the DHA-derived 10,17S-docosatriene NPD1 (neuroprotectin D1), a tandem brain cytosolic phospholipase A(2) and 15-lipoxygenase enzymatic system that biosynthesizes NPD1, and a small family of anti-apoptotic neuroprotective genes that encode Bcl-2, Bcl-X(L) and Bfl-1 (A1). This paper reviews current ideas regarding DHA and the oxygenated DHA derivative NPD1, intrinsically triggered biolipid neuroprotectants that along with their associated rescue pathways, contribute to life-or-death decisions of brain cells during homoeostasis, aging and neurodegenerative disease.


Subject(s)
Alzheimer Disease/pathology , Cell Survival/physiology , Serum Amyloid A Protein/physiology , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Apoptosis , Brain/pathology , Brain/physiopathology , Cell Survival/drug effects , Humans , Models, Neurological , Multigene Family , Nerve Growth Factors/pharmacology , Neurotoxins , Signal Transduction
17.
Med Hypotheses ; 64(2): 320-7, 2005.
Article in English | MEDLINE | ID: mdl-15607565

ABSTRACT

Herpes simplex virus type 1 (HSV-1) is latent in the nervous system of most humans. Ball [Can J Neurol Sci 9 (1982) 303] first suggested the hypothesis that HSV-1 could be involved in the pathogenesis of Alzheimer's Disease (AD) by noting that regions of the brain particularly and earliest affected in AD were the same as those most damaged during HSV encephalitis. Data from Itzhaki's research suggests that HSV-1 in the brain and the carriage of an apolipoprotein E allele 4 (ApoE e4) together confer risk for AD [J Pathol 97 (2002) 395], [Mol Chem Neuropathol 28 (1996) 135], [Alzheimer's Rep 1 (1998) 173], [Biochem Soc Trans 26 (1998) 273]. Of the two other studies based on Itzhaki's findings, one showed similar results [Lancet 349 (1997) 1102], and the other showed a similar trend [Lancet 351 (1998) 1330], [Lancet 352 (1998) 1312]. To further examine the role of HSV-1 in the etiology of AD, we have formulated a Neuroinvasive Score that quantifies the presence and viral load of HSV-1 in eight brain regions. These regions are: entorhinal cortex, hippocampus, pons, cerebellum, and neocortex (temporal, parietal, occipital, and frontal). We hypothesize that the Neuroinvasive Score that encompasses the presence, amount, and extent of HSV-1 spreading (neuroinvasiveness), will correlate with the genetic risk factor, ApoE e4, in the assessment of autopsy samples from AD patients. If the neuroinvasive score can be directly correlated to the different stages of AD (mild, moderate, severe), this will strengthen the hypothesis that HSV-1 is involved in AD and that ApoE e4 also confers risk for the development and progression of AD.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/virology , Brain/virology , Herpesvirus 1, Human , Viral Load/methods , Humans , Risk Factors
18.
Mol Psychiatry ; 7(8): 891-8, 2002.
Article in English | MEDLINE | ID: mdl-12232783

ABSTRACT

We have analyzed the 5'-upstream promoter region of the presenilin 2 gene (PSEN2) for regulatory elements and examined Alzheimer disease (AD) patients and non-demented individuals for polymorphisms in the 5' upstream promoter region of the PSEN2 gene. Direct sequencing analysis detected a common single adenine (A) nucleotide deletion polymorphism in the upstream promoter region of the PSEN2 gene. Examination of cohorts of AD patients and age-matched control individuals revealed no statistically significant differences in the frequency of this polymorphism when compared with the total sample of AD patients and control individuals. However, subgroup and regression analysis suggested that the relatively rare -A/-A genotype increases risk of AD among subjects lacking apolipoprotein E (APOE) epsilon4 and among persons ages 65 years and younger. DNA sequence and DNA-protein binding analysis demonstrated that this mutation negates binding with putative repressor transcription factor (TF), interferon regulatory factor 2 (IRF2), in nuclear extracts prepared from the aged human brain neocortex. However this mutation creates a potential regulatory element, C/EBPbeta, that is responsive to pro-inflammatory (PI) induction. The expression activity assay with luciferase reporter gene into normal human neural progenitor cells in primary culture shows that the mutant PSEN2 regulatory region exhibits a 1.8-fold higher level of basal expression and is sensitive to IL-1beta and Abeta42, but that it is synergistically induced 3.2-fold over the wild-type PSEN2 by [IL-1beta+Abeta42]. These results suggest that under Pl and oxygen stress conditions relatively minor variations in PSEN2 promoter DNA sequence structure can enhance PSEN2 gene expression and that consequently these may play a role in the induction and/or proliferation of a Pl response in AD brain.


Subject(s)
Alzheimer Disease/genetics , Membrane Proteins/genetics , 5' Untranslated Regions/genetics , Aged , Alzheimer Disease/epidemiology , Alzheimer Disease/metabolism , Cells, Cultured , DNA Mutational Analysis , Electrophoretic Mobility Shift Assay , Genes, Reporter , Genetic Predisposition to Disease/epidemiology , Genetic Variation , Humans , Luciferases/genetics , Middle Aged , Oxidative Stress/genetics , Polymorphism, Genetic , Presenilin-2 , Promoter Regions, Genetic , Risk Factors , Stem Cells/cytology , Transcription Factors/metabolism
19.
Brain Res ; 894(1): 37-45, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11245813

ABSTRACT

Metallothionein III (MT-III) is a functionally distinct member of the metallothionein family that displays neuroinhibitory activity and is involved in the repair of neuronal damage. Altered expression levels of MT-III have been observed in Alzheimer's disease (AD) which has led to suggestions that it could be a mitigating factor in AD-related neuronal dysfunction. However, conflicting results have been reported on this issue which may be due to methodological differences and/or sampling size. In the current study, we have assessed MT-III expression in a large number of AD cases through the quantification of mRNA as well as by immunohistochemistry and Western blotting using an MT-III specific antibody. The results of this comprehensive study indicate that the mononucleosome DNA encoding MT-III is occluded preventing transcription and that message levels are reduced by approximately 30%. In addition, protein levels were specifically decreased by approximately 55% in temporal cortex. These data support the conclusion that MT-III is significantly downregulated in AD and may contribute to the loss of its protective effects and/or repair functions that lead to an exacerbation of the pathogenic processes.


Subject(s)
Alzheimer Disease/metabolism , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism , Alzheimer Disease/pathology , Frontal Lobe/metabolism , Humans , Metallothionein 3 , Nucleosomes/metabolism , Temporal Lobe/metabolism
20.
Neuroreport ; 12(1): 53-7, 2001 Jan 22.
Article in English | MEDLINE | ID: mdl-11201091

ABSTRACT

Presenilin-2 (PS2; AD4), a regulator of intercellular signaling during CNS development and cell fate determination, appears to be involved in pathogenic processing of beta-amyloid precursor protein (betaAPP) into potentially neurotoxic beta-amyloid (Abeta) peptides. The PS2 gene promoter contains multiple DNA binding sites for the relatively rare hypoxia-inducible transcription factor HIF-1, suggesting that PS2 expression may be a sensitive indicator of decreased oxygen availability. We have used a cycled hypoxia/hyperoxia (10-50% O2) protocol followed by normoxia (20% O2) as a retinal model of retinopathy of prematurity to induce neovascularization (NV) in rat pups. Retinal cell nuclear extracts from pups undergoing hypoxia exhibited a dramatic increase in HIF-1-DNA binding, followed by a delayed (2-7 day) elevation of PS2 RNA message and protein. PS2 gene activation during hypoxia may direct cellular fate towards pathoangiogenesis and intercellular PS2-mediated signaling dysfunction.


Subject(s)
DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression/physiology , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Retinal Neovascularization/metabolism , Retinopathy of Prematurity/metabolism , Transcription Factors , Animals , Animals, Newborn , Female , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Infant, Newborn , Pregnancy , Presenilin-2 , Rats , Retina/metabolism , Retinal Neovascularization/etiology , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...