Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39026803

ABSTRACT

Neurons and glia work together to dynamically regulate neural circuit assembly and maintenance. In this study, we show Drosophila exhibit large-scale synapse formation and elimination as part of normal CNS circuit maturation, and that glia use conserved molecules to regulate these processes. Using a high throughput ELISA-based in vivo screening assay, we identify new glial genes that regulate synapse numbers in Drosophila in vivo, including the scavenger receptor ortholog Croquemort (Crq). Crq acts as an essential regulator of glial-dependent synapse elimination during development, with glial Crq loss leading to excess CNS synapses and progressive seizure susceptibility in adults. Loss of Crq in glia also prevents age-related synaptic loss in the adult brain. This work provides new insights into the cellular and molecular mechanisms that underlie synapse development and maintenance across the lifespan, and identifies glial Crq as a key regulator of these processes.

2.
Neuron ; 112(1): 93-112.e10, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38096817

ABSTRACT

Astrocytes play crucial roles in regulating neural circuit function by forming a dense network of synapse-associated membrane specializations, but signaling pathways regulating astrocyte morphogenesis remain poorly defined. Here, we show the Drosophila lipid-binding G protein-coupled receptor (GPCR) Tre1 is required for astrocytes to establish their intricate morphology in vivo. The lipid phosphate phosphatases Wunen/Wunen2 also regulate astrocyte morphology and, via Tre1, mediate astrocyte-astrocyte competition for growth-promoting lipids. Loss of s1pr1, the functional analog of Tre1 in zebrafish, disrupts astrocyte process elaboration, and live imaging and pharmacology demonstrate that S1pr1 balances proper astrocyte process extension/retraction dynamics during growth. Loss of Tre1 in flies or S1pr1 in zebrafish results in defects in simple assays of motor behavior. Tre1 and S1pr1 are thus potent evolutionarily conserved regulators of the elaboration of astrocyte morphological complexity and, ultimately, astrocyte control of behavior.


Subject(s)
Drosophila Proteins , Zebrafish , Animals , Astrocytes/metabolism , Drosophila/metabolism , Drosophila Proteins/metabolism , Phospholipids/metabolism , Receptors, G-Protein-Coupled/metabolism , Sphingosine-1-Phosphate Receptors/metabolism
3.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Article in English | MEDLINE | ID: mdl-34686600

ABSTRACT

Drosophila is a powerful model in which to perform genetic screens, but screening assays that are both rapid and can be used to examine a wide variety of cellular and molecular pathways are limited. Drosophila offer an extensive toolbox of GFP-based transcriptional reporters, GFP-tagged proteins, and driver lines, which can be used to express GFP in numerous subpopulations of cells. Thus, a tool that can rapidly and quantitatively evaluate GFP levels in Drosophila tissue would provide a broadly applicable screening platform. We developed a GFP-based enzyme-linked immunosorbent assay (ELISA) that can detect GFP in Drosophila lysates collected from whole animals and dissected tissues across all stages of Drosophila development. We demonstrate that this assay can detect membrane-localized GFP in a variety of neuronal and glial populations and validate that it can identify genes that change the morphology of these cells, as well as changes in STAT and JNK transcriptional activity. We found that this assay can detect endogenously GFP-tagged proteins, including Draper, Cryptochrome, and the synaptic marker Brp. This approach is able to detect changes in Brp-GFP signal during developmental synaptic remodeling, and known genetic regulators of glial synaptic engulfment could be identified using this ELISA method. Finally, we used the assay to perform a small-scale screen, which identified Syntaxins as potential regulators of astrocyte-mediated synapse elimination. Together, these studies establish an ELISA as a rapid, easy, and quantitative in vivo screening method that can be used to assay a wide breadth of fundamental biological questions.


Subject(s)
Drosophila/genetics , Enzyme-Linked Immunosorbent Assay/methods , Animals , Green Fluorescent Proteins/genetics , Neuroglia/metabolism , Reproducibility of Results
4.
Neurobiol Dis ; 145: 105072, 2020 11.
Article in English | MEDLINE | ID: mdl-32890775

ABSTRACT

Genetic studies identified mutations in several immune-related genes that confer increased risk for developing Alzheimer's disease (AD), suggesting a key role for microglia in AD pathology. Microglia are recruited to and actively modulate the local toxicity of amyloid plaques in models of AD through these cells' transcriptional and functional reprogramming to a disease-associated phenotype. However, it remains unknown whether microglia actively respond to amyloid accumulation before plaque deposition in AD. We compared microglial interactions with neurons that exhibit amyloid accumulation to those that do not in 1-month-old 5XFAD mice to determine which aspects of microglial morphology and function are altered by early 6E10+ amyloid accumulation. We provide evidence of preferential microglial process engagement of amyloid laden neurons. Microglia, on exposure to amyloid, also increase their internalization of neurites even before plaque onset. Unexpectedly, we found that triggering receptor expressed on myeloid cells 2 (TREM2), which is critical for microglial responses to amyloid plaque pathology later in disease, is not required for enhanced microglial interactions with neurons or neurite internalization early in disease. However, TREM2 was still required for early morphological changes exhibited by microglia. These data demonstrate that microglia sense and respond to amyloid accumulation before plaques form using a distinct mechanism from the TREM2-dependent pathway required later in disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Membrane Glycoproteins/metabolism , Microglia/metabolism , Neurons/metabolism , Plaque, Amyloid/metabolism , Receptors, Immunologic/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Microglia/pathology , Neurons/pathology , Plaque, Amyloid/pathology
5.
Glia ; 67(10): 1873-1892, 2019 10.
Article in English | MEDLINE | ID: mdl-31265185

ABSTRACT

Variants in the microglial receptor TREM2 confer risk for multiple neurodegenerative diseases. However, it remains unknown how this receptor functions on microglia to modulate these diverse neuropathologies. To understand the role of TREM2 on microglia more generally, we investigated changes in microglial function in Trem2-/- mice. We found that loss of TREM2 impairs normal neurodevelopment, resulting in reduced synapse number across the cortex and hippocampus in 1-month-old mice. This reduction in synapse number was not due directly to alterations in interactions between microglia and synapses. Rather, TREM2 was required for microglia to limit synaptic engulfment by astrocytes during development. While these changes were largely normalized later in adulthood, high fat diet administration was sufficient to reinitiate TREM2-dependent modulation of synapse loss. Together, this identifies a novel role for microglia in instructing synaptic pruning by astrocytes to broadly regulate appropriate synaptic refinement, and suggests novel candidate mechanisms for how TREM2 and microglia could influence synaptic loss in brain injury and disease.


Subject(s)
Astrocytes/metabolism , Brain/growth & development , Brain/metabolism , Membrane Glycoproteins/metabolism , Microglia/metabolism , Receptors, Immunologic/metabolism , Synapses/metabolism , Animals , Diet, High-Fat/adverse effects , Female , Male , Membrane Glycoproteins/genetics , Mice, Knockout , Random Allocation , Receptors, Immunologic/genetics , Tissue Culture Techniques
6.
Mol Neurodegener ; 13(1): 29, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29859094

ABSTRACT

BACKGROUND: The R47H variant of Triggering Receptor Expressed on Myeloid cells 2 (TREM2) confers greatly increased risk for Alzheimer's disease (AD), reflective of a central role for myeloid cells in neurodegeneration. Understanding how this variant confers AD risk promises to provide important insights into how myeloid cells contribute to AD pathogenesis and progression. METHODS: In order to investigate this mechanism, CRISPR/Cas9 was used to generate a mouse model of AD harboring one copy of the single nucleotide polymorphism (SNP) encoding the R47H variant in murine Trem2. TREM2 expression, myeloid cell responses to amyloid deposition, plaque burden, and neuritic dystrophy were assessed at 4 months of age. RESULTS: AD mice heterozygous for the Trem2 R47H allele exhibited reduced total Trem2 mRNA expression, reduced TREM2 expression around plaques, and reduced association of myeloid cells with plaques. These results were comparable to AD mice lacking one copy of Trem2. AD mice heterozygous for the Trem2 R47H allele also showed reduced myeloid cell responses to amyloid deposition, including a reduction in proliferation and a reduction in CD45 expression around plaques. Expression of the Trem2 R47H variant also reduced dense core plaque number but increased plaque-associated neuritic dystrophy. CONCLUSIONS: These data suggest that the AD-associated TREM2 R47H variant increases risk for AD by conferring a loss of TREM2 function and enhancing neuritic dystrophy around plaques.


Subject(s)
Alzheimer Disease , Brain/pathology , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Animals , Membrane Glycoproteins/genetics , Mice , Phenotype , Polymorphism, Single Nucleotide , Receptors, Immunologic/genetics
7.
Mol Neurodegener ; 12(1): 56, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28768545

ABSTRACT

TREM2 variants have been identified as risk factors for Alzheimer's disease (AD) and other neurodegenerative diseases (NDDs). Because TREM2 encodes a receptor exclusively expressed on immune cells, identification of these variants conclusively demonstrates that the immune response can play an active role in the pathogenesis of NDDs. These TREM2 variants also confer the highest risk for developing Alzheimer's disease of any risk factor identified in nearly two decades, suggesting that understanding more about TREM2 function could provide key insights into NDD pathology and provide avenues for novel immune-related NDD biomarkers and therapeutics. The expression, signaling and function of TREM2 in NDDs have been extensively investigated in an effort to understand the role of immune function in disease pathogenesis and progression. We provide a comprehensive review of our current understanding of TREM2 biology, including new insights into the regulation of TREM2 expression, and TREM2 signaling and function across NDDs. While many open questions remain, the current body of literature provides clarity on several issues. While it is still often cited that TREM2 expression is decreased by pro-inflammatory stimuli, it is now clear that this is true in vitro, but inflammatory stimuli in vivo almost universally increase TREM2 expression. Likewise, while TREM2 function is classically described as promoting an anti-inflammatory phenotype, more than half of published studies demonstrate a pro-inflammatory role for TREM2, suggesting that its role in inflammation is much more complex. Finally, these components of TREM2 biology are applied to a discussion of how TREM2 impacts NDD pathologies and the latest assessment of how these findings might be applied to immune-directed clinical biomarkers and therapeutics.


Subject(s)
Biomarkers/metabolism , Membrane Glycoproteins/metabolism , Microglia/metabolism , Neurodegenerative Diseases/metabolism , Receptors, Immunologic/metabolism , Animals , Disease Models, Animal , Disease Progression , Humans , Membrane Glycoproteins/genetics , Neurodegenerative Diseases/genetics , Receptors, Immunologic/genetics
8.
J Neurosci ; 37(3): 637-647, 2017 01 18.
Article in English | MEDLINE | ID: mdl-28100745

ABSTRACT

Neuroinflammation is an important contributor to Alzheimer's disease (AD) pathogenesis, as underscored by the recent identification of immune-related genetic risk factors for AD, including coding variants in the gene TREM2 (triggering receptor expressed on myeloid cells 2). Understanding TREM2 function promises to provide important insights into how neuroinflammation contributes to AD pathology. However, studies so far have produced seemingly conflicting results, with reports that amyloid pathology can be both decreased and increased in TREM2-deficient AD mouse models. In this study, we unify these previous findings by demonstrating that TREM2 deficiency ameliorates amyloid pathology early, but exacerbates it late in disease progression in the APPPS1-21 mouse model of AD. We also demonstrate that TREM2 deficiency decreases plaque-associated myeloid cell accumulation by reducing cell proliferation, specifically late in pathology. In addition, TREM2 deficiency reduces myeloid cell internalization of amyloid throughout pathology, but decreases inflammation-related gene transcript levels selectively late in disease progression. Together, these results suggest that TREM2 plays distinct functional roles at different stages in AD pathology. SIGNIFICANCE STATEMENT: Alzheimer's disease (AD) is a devastating neurodegenerative disorder and there are currently no effective treatments that modify disease progression. However, the recent identification of genetic risk factors for AD promises to provide new insight into AD biology and possible new therapeutic targets. Among these risk factors, variants in the gene TREM2 (triggering receptor expressed on myeloid cells 2) confer greatly elevated risk for developing the disease. We demonstrate that TREM2 deficiency has opposing effects on AD-related pathologies at early and late stages of disease progression, unifying previous work in the field. In addition, we examine how TREM2 affects the function of the brain immune cell populations in which it is expressed throughout disease progression to understand possible mechanisms underlying its differential impacts on pathology.


Subject(s)
Alzheimer Disease/metabolism , Disease Models, Animal , Disease Progression , Membrane Glycoproteins/deficiency , Receptors, Immunologic/deficiency , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Female , Humans , Male , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Receptors, Immunologic/genetics
9.
J Neurovirol ; 23(1): 47-66, 2017 02.
Article in English | MEDLINE | ID: mdl-27873219

ABSTRACT

The major reservoirs for HIV in the CNS are in the microglia, perivascular macrophages, and to a lesser extent, astrocytes. To study the molecular events controlling HIV expression in the microglia, we developed a reliable and robust method to immortalize microglial cells from primary glia from fresh CNS tissues and commercially available frozen glial cells. Primary human cells, including cells obtained from adult brain tissue, were transformed with lentiviral vectors expressing SV40 T antigen or a combination of SVR40 T antigen and hTERT. The immortalized cells have microglia-like morphology and express key microglial surface markers including CD11b, TGFßR, and P2RY12. Importantly, these cells were confirmed to be of human origin by sequencing. The RNA expression profiles identified by RNA-seq are also characteristic of microglial cells. Furthermore, the cells demonstrate the expected migratory and phagocytic activity, and the capacity to mount an inflammatory response characteristic of primary microglia. The immortalization method has also been successfully applied to a wide range of microglia from other species (macaque, rat, and mouse). To investigate different aspects of HIV molecular regulation in CNS, the cells have been superinfected with HIV reporter viruses and latently infected clones have been selected that reactive HIV in response to inflammatory signals. The cell lines we have developed and rigorously characterized will provide an invaluable resource for the study of HIV infection in microglial cells as well as studies of microglial cell function.


Subject(s)
Antigens, Viral, Tumor/genetics , Founder Effect , Microglia/pathology , Transformation, Genetic , Adult , Animals , Antigens, Viral, Tumor/metabolism , Biomarkers/metabolism , Brain/metabolism , Brain/pathology , Brain/virology , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cell Movement , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Macaca , Mice , Microglia/metabolism , Microglia/virology , Phagocytosis , Primary Cell Culture , Rats , Receptors, Purinergic P2Y12/genetics , Receptors, Purinergic P2Y12/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism
11.
J Neurosci ; 35(16): 6532-43, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904803

ABSTRACT

Alzheimer's disease (AD) is characterized by a robust inflammatory response elicited by the accumulation and subsequent deposition of amyloid (Aß) within the brain. The brain's immune cells migrate to and invest their processes within Aß plaques but are unable to efficiently phagocytose and clear plaques from the brain. Previous studies have shown that treatment of myeloid cells with nuclear receptor agonists increases expression of phagocytosis-related genes. In this study, we elucidate a novel mechanism by which nuclear receptors act to enhance phagocytosis in the AD brain. Treatment of murine models of AD with agonists of the nuclear receptors PPARγ, PPARδ, LXR, and RXR stimulated microglial phagocytosis in vitro and rapidly induced the expression of the phagocytic receptors Axl and MerTK. In murine models of AD, we found that plaque-associated macrophages expressed Axl and MerTK and treatment of the cells with an RXR agonist further induced their expression, coincident with the rapid reduction in plaque burden. Further characterization of MerTK(+)/Axl(+) macrophages revealed that they also expressed the phagocytic receptor TREM2 and high levels of CD45, consistent with a peripheral origin of these cells. Importantly, in an ex vivo slice assay, nuclear receptor agonist treatment reversed the AD-related suppression of phagocytosis through a MerTK-dependent mechanism. Thus, nuclear receptor agonists increase MerTK and Axl expression on plaque-associated immune cells, consequently licensing their phagocytic activity and promoting plaque clearance.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Disease Models, Animal , Membrane Glycoproteins/metabolism , Myeloid Cells/metabolism , Phagocytosis/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Immunologic/metabolism , Animals , Benzoates/pharmacology , Benzylamines/pharmacology , Bexarotene , Cells, Cultured , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Leukocyte Common Antigens/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Microglia/drug effects , Microglia/metabolism , Myeloid Cells/drug effects , Phagocytosis/drug effects , Pioglitazone , Plaque, Amyloid/metabolism , Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Cytoplasmic and Nuclear/agonists , Tetrahydronaphthalenes/pharmacology , Thiazoles/pharmacology , Thiazolidinediones/pharmacology , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
12.
J Exp Med ; 212(3): 287-95, 2015 Mar 09.
Article in English | MEDLINE | ID: mdl-25732305

ABSTRACT

Variants in triggering receptor expressed on myeloid cells 2 (TREM2) confer high risk for Alzheimer's disease (AD) and other neurodegenerative diseases. However, the cell types and mechanisms underlying TREM2's involvement in neurodegeneration remain to be established. Here, we report that TREM2 is up-regulated on myeloid cells surrounding amyloid deposits in AD mouse models and human AD tissue. TREM2 was detected on CD45(hi)Ly6C(+) myeloid cells, but not on P2RY12(+) parenchymal microglia. In AD mice deficient for TREM2, the CD45(hi)Ly6C(+) macrophages are virtually eliminated, resulting in reduced inflammation and ameliorated amyloid and tau pathologies. These data suggest a functionally important role for TREM2(+) macrophages in AD pathogenesis and an unexpected, detrimental role of TREM2 in AD pathology. These findings have direct implications for future development of TREM2-targeted therapeutics.


Subject(s)
Alzheimer Disease/pathology , Macrophages/metabolism , Macrophages/pathology , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Age Factors , Aged , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Leukocyte Common Antigens/metabolism , Male , Membrane Glycoproteins/genetics , Mice, Transgenic , Receptors, Immunologic/genetics , Up-Regulation , tau Proteins/metabolism
13.
Neurotherapeutics ; 12(1): 81-93, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25404051

ABSTRACT

Alzheimer's disease (AD) is typified by a robust microglial-mediated inflammatory response within the brain. Indeed, microglial accumulation around plaques in AD is one of the classical hallmarks of the disease pathology. Although microglia have the capacity to remove ß-amyloid deposits and alleviate disease pathology, they fail to do so. Instead, they become chronically activated and promote inflammation-mediated impairment of cognition and cytotoxicity. However, if microglial function could be altered to engage their phagocytic response, promote their tissue maintenance functions, and prevent release of factors that promote tissue damage, this could provide therapeutic benefit. This review is focused on the current knowledge of microglial homeostatic mechanisms in AD, and mechanisms involved in the regulation of microglial phenotype in this context.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Microglia/metabolism , Animals , Humans , Phagocytosis/physiology
14.
J Neurosci ; 34(37): 12538-46, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-25209291

ABSTRACT

Several Alzheimer's disease (AD) risk genes are specifically expressed by microglia within the CNS. However, the mechanisms by which microglia regulate the pathological hallmarks of AD--extracellular deposition of ß-amyloid (Aß) and intraneuronal hyperphosphorylation of microtubule-associated protein tau (MAPT)--remain to be established. Notably, deficiency for the microglial CX3CR1 receptor has opposing effects on Aß and MAPT pathologies. CX3CL1, the neuronally derived cognate ligand for CX3CR1, signals both in membrane-anchored and soluble forms. In this study, we sought to determine the relative contribution on membrane-anchored versus soluble CX3CL1 in regulating the microglia-mediated amelioration of Aß pathology, as well as provide insight into the potential downstream microglial-based mechanisms. As expected, CX3CL1 deficiency reduced Aß deposition in APPPS1 animals in a similar manner to CX3CR1 deficiency. Surprisingly, however, CX3CL1-deficient APPPS1 animals exhibited enhanced neuronal MAPT phosphorylation despite reduced amyloid burden. Importantly, neither of these phenotypes was altered by transgenic expression of the soluble CX3CL1 isoform, suggesting that it is the membrane-anchored version of CX3CL1 that regulates microglial phagocytosis of Aß and neuronal MAPT phosphorylation. Analysis of transcript levels in purified microglia isolated from APPPS1 mice with the various CX3CL1/CX3CR1 genotypes revealed increased expression of inflammatory cytokines and phagocytic markers, which was associated with activation of p38 mitogen-activated protein kinase and Aß internalization within microglia. Together, these studies challenge the "frustrated phagocytosis" concept and suggest that neuronal-microglial communication link the two central AD pathologies.


Subject(s)
Amyloid beta-Peptides/metabolism , Cell Membrane/metabolism , Chemokine CXCL1/metabolism , MAP Kinase Signaling System , Microglia/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , tau Proteins/metabolism , Alzheimer Disease , Animals , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
15.
Analyst ; 137(9): 2158-65, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22416270

ABSTRACT

Histamine is among the most poorly understood biogenic amines, yet the histaminergic system spreads throughout the brain and has been implicated in functions as diverse as homeostasis and synaptic plasticity. Not surprisingly then, it has been linked to a number of conditions including minimally conscious state, persistent vegetative state, epilepsy, addiction, cluster headache, essential tremor, and Parkinson's disease. We have previously reported that the Wireless Instantaneous Neurotransmitter Concentration Sensing (WINCS) system can monitor dopamine, serotonin, and adenosine using fast-scan cyclic voltammetry (FSCV). Here, we demonstrate the expanded capability of the WINCS system to measure histamine. The optimal FSCV waveform was determined to be a triangle wave scanned between -0.4 and +1.4 V at a rate of 400 V s(-1) applied at 10 Hz. Using this optimized FSCV parameter, we found histamine release was induced by high frequency electrical stimulation at the tuberomammillary nucleus in rat brain slices. Our results suggest that the WINCS system can provide reliable, high fidelity measurements of histamine, consistently showing oxidative currents at +1.3 V, a finding that may have important clinical implications.


Subject(s)
Electrochemistry/methods , Histamine/analysis , Neurotransmitter Agents/analysis , Wireless Technology , Animals , Electric Stimulation , Hypothalamic Area, Lateral/cytology , Male , Rats , Rats, Sprague-Dawley , Time Factors
16.
PLoS One ; 5(4): e10296, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20421971

ABSTRACT

BACKGROUND: The present study sought to further investigate the in vitro and in vivo anticancer effects of a representative omega-3 fatty acid, docosahexaenoic acid (DHA), with a focus on assessing the induction of oxidative stress and apoptosis as an important mechanism for its anticancer actions. METHODOLOGY/PRINCIPAL FINDINGS: In vitro studies showed that DHA strongly reduces the viability and DNA synthesis of MCF-7 human breast cancer cells in culture, and also promotes cell death via apoptosis. Mechanistically, accumulation of reactive oxygen species and activation of caspase 8 contribute critically to the induction of apoptotic cell death. Co-presence of antioxidants or selective inhibition or knockdown of caspase 8 each effectively abrogates the cytotoxic effect of DHA. Using athymic nude mice as an in vivo model, we found that feeding animals the 5% fish oil-supplemented diet for 6 weeks significantly reduces the growth of MCF-7 human breast cancer cells in vivo through inhibition of cancer cell proliferation as well as promotion of cell death. Using 3-nitrotyrosine as a parameter, we confirmed that the fish oil-supplemented diet significantly increases oxidative stress in tumor cells in vivo. Analysis of fatty acid content in plasma and tissues showed that feeding animals a 5% fish oil diet increases the levels of DHA and eicosapentaenoic acid in both normal and tumorous mammary tissues by 329% and 300%, respectively. CONCLUSIONS/SIGNIFICANCE: DHA can strongly induce apoptosis in human MCF-7 breast cancer cells both in vitro and in vivo. The induction of apoptosis in these cells is selectively mediated via caspase 8 activation. These observations call for further studies to assess the effectiveness of fish oil as a dietary supplement in the prevention and treatment of human breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Caspase 8/metabolism , Docosahexaenoic Acids/pharmacology , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Agents , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Fatty Acids, Omega-3/pharmacology , Female , Fish Oils/administration & dosage , Fish Oils/therapeutic use , Humans , Mice , Mice, Nude , Oxidative Stress , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...