Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(33): e2121040119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35943986

ABSTRACT

Regulation of firing rate homeostasis constitutes a fundamental property of central neural circuits. While intracellular Ca2+ has long been hypothesized to be a feedback control signal, the molecular machinery enabling a network-wide homeostatic response remains largely unknown. We show that deletion of insulin-like growth factor-1 receptor (IGF-1R) limits firing rate homeostasis in response to inactivity, without altering the distribution of baseline firing rates. The deficient firing rate homeostatic response was due to disruption of both postsynaptic and intrinsic plasticity. At the cellular level, we detected a fraction of IGF-1Rs in mitochondria, colocalized with the mitochondrial calcium uniporter complex (MCUc). IGF-1R deletion suppressed transcription of the MCUc members and burst-evoked mitochondrial Ca2+ (mitoCa2+) by weakening mitochondria-to-cytosol Ca2+ coupling. Overexpression of either mitochondria-targeted IGF-1R or MCUc in IGF-1R-deficient neurons was sufficient to rescue the deficits in burst-to-mitoCa2+ coupling and firing rate homeostasis. Our findings indicate that mitochondrial IGF-1R is a key regulator of the integrated homeostatic response by tuning the reliability of burst transfer by MCUc. Based on these results, we propose that MCUc acts as a homeostatic Ca2+ sensor. Faulty activation of MCUc may drive dysregulation of firing rate homeostasis in aging and in brain disorders associated with aberrant IGF-1R/MCUc signaling.


Subject(s)
Calcium Channels , Calcium , Receptor, IGF Type 1 , Animals , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Gene Deletion , Homeostasis , Mice , Neuronal Plasticity , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Reproducibility of Results
2.
Endocrinology ; 158(7): 2269-2283, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28881863

ABSTRACT

Growth hormone (GH) and insulinlike growth factor (IGF) promote aging and age-related pathologies. Inhibiting this pathway by targeting IGF receptor (IGF-1R) is a promising strategy to extend life span, alleviate age-related diseases, and reduce tumor growth. Although anti-IGF-1R agents are being developed, long-term effects of IGF-1R blockade remain unknown. In this study, we used ubiquitous inducible IGF-1R knockout (UBIKOR) to suppress signaling in all adult tissues and screened health extensively. Surprisingly, UBIKOR mice showed no overt defects and presented with rather inconspicuous health, including normal cognition. Endocrine GH and IGF-1 were strongly upregulated without causing acromegaly. UBIKOR mice were strikingly lean with coordinate changes in body composition and organ size. They were insulin resistant but preserved physiological energy expenditure and displayed enhanced fasting metabolic flexibility. Thus, long-term IGF-1R blockade generated beneficial effects on aging-relevant metabolism, but exposed to high GH. This needs to be considered when targeting IGF-1R to protect from neurodegeneration, retard aging, or fight cancer.


Subject(s)
Energy Metabolism/genetics , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/genetics , Thinness/genetics , Animals , Body Composition/drug effects , Body Composition/genetics , Energy Metabolism/drug effects , Female , Gene Deletion , Human Growth Hormone/analogs & derivatives , Human Growth Hormone/pharmacology , Insulin Resistance/genetics , Insulin-Like Growth Factor I/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, IGF Type 1/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/genetics , Thinness/metabolism
3.
Proc Natl Acad Sci U S A ; 114(36): 9731-9736, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827363

ABSTRACT

When food resources are scarce, endothermic animals can lower core body temperature (Tb). This phenomenon is believed to be part of an adaptive mechanism that may have evolved to conserve energy until more food becomes available. Here, we found in the mouse that the insulin-like growth factor 1 receptor (IGF-1R) controls this response in the central nervous system. Pharmacological or genetic inhibition of IGF-1R enhanced the reduction of temperature and of energy expenditure during calorie restriction. Full blockade of IGF-1R affected female and male mice similarly. In contrast, genetic IGF-1R dosage was effective only in females, where it also induced transient and estrus-specific hypothermia in animals fed ad libitum. These effects were regulated in the brain, as only central, not peripheral, pharmacological activation of IGF-1R prevented hypothermia during calorie restriction. Targeted IGF-1R knockout selectively in forebrain neurons revealed that IGF signaling also modulates calorie restriction-dependent Tb regulation in regions rostral of the canonical hypothalamic nuclei involved in controlling body temperature. In aggregate, these data identify central IGF-1R as a mediator of the integration of nutrient and temperature homeostasis. They also show that calorie restriction, IGF-1R signaling, and body temperature, three of the main regulators of metabolism, aging, and longevity, are components of the same pathway.


Subject(s)
Caloric Restriction/adverse effects , Hypothermia/physiopathology , Receptor, IGF Type 1/physiology , Aging/physiology , Animals , Energy Metabolism/physiology , Female , Gene Dosage , Homeostasis/physiology , Hypothermia/etiology , Hypothermia/prevention & control , Longevity/physiology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics , Sex Characteristics , Signal Transduction/physiology
4.
Brain ; 140(7): 2012-2027, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28595357

ABSTRACT

Seminal studies using post-mortem brains of patients with Alzheimer's disease evidenced aberrant insulin-like growth factor 1 receptor (IGF1R) signalling. Addressing causality, work in animal models recently demonstrated that long-term suppression of IGF1R signalling alleviates Alzheimer's disease progression and promotes neuroprotection. However, the underlying mechanisms remain largely elusive. Here, we showed that genetically ablating IGF1R in neurons of the ageing brain efficiently protects from neuroinflammation, anxiety and memory impairments induced by intracerebroventricular injection of amyloid-ß oligomers. In our mutant mice, the suppression of IGF1R signalling also invariably led to small neuronal soma size, indicative of profound changes in cellular homeodynamics. To gain insight into transcriptional signatures leading to Alzheimer's disease-relevant neuronal defence, we performed genome-wide microarray analysis on laser-dissected hippocampal CA1 after neuronal IGF1R knockout, in the presence or absence of APP/PS1 transgenes. Functional analysis comparing neurons in early-stage Alzheimer's disease with IGF1R knockout neurons revealed strongly convergent transcriptomic signatures, notably involving neurite growth, cytoskeleton organization, cellular stress response and neurotransmission. Moreover, in Alzheimer's disease neurons, a high proportion of genes responding to Alzheimer's disease showed a reversed differential expression when IGF1R was deleted. One of the genes consistently highlighted in genome-wide comparison was the neurofilament medium polypeptide Nefm. We found that NEFM accumulated in hippocampus in the presence of amyloid pathology, and decreased to control levels under IGF1R deletion, suggesting that reorganized cytoskeleton likely plays a role in neuroprotection. These findings demonstrated that significant resistance of the brain to amyloid-ß can be achieved lifelong by suppressing neuronal IGF1R and identified IGF-dependent molecular pathways that coordinate an intrinsic program for neuroprotection against proteotoxicity. Our data also indicate that neuronal defences against Alzheimer's disease rely on an endogenous gene expression profile similar to the neuroprotective response activated by genetic disruption of IGF1R signalling. This study highlights neuronal IGF1R signalling as a relevant target for developing Alzheimer's disease prevention strategies.


Subject(s)
Alzheimer Disease/metabolism , CA1 Region, Hippocampal/metabolism , Neuroprotective Agents/metabolism , Receptor, IGF Type 1/deficiency , Receptor, IGF Type 1/genetics , Transcriptome , Aging/metabolism , Alzheimer Disease/complications , Amyloid beta-Peptides/administration & dosage , Animals , Anxiety/chemically induced , Anxiety/complications , Anxiety/prevention & control , Encephalitis/chemically induced , Encephalitis/complications , Encephalitis/prevention & control , Female , Infusions, Intraventricular , Male , Memory Disorders/chemically induced , Memory Disorders/complications , Memory Disorders/prevention & control , Mice , Mice, Knockout , Mice, Transgenic , Neurons/metabolism
5.
J Cereb Blood Flow Metab ; 37(2): 396-412, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26762506

ABSTRACT

Insulin-like growth factors control numerous processes, namely somatic growth, metabolism and stress resistance, connecting this pathway to aging and age-related diseases. Insulin-like growth factor signaling also impacts on neurogenesis, neuronal survival and structural plasticity. Recent reports demonstrated that diminished insulin-like growth factor signaling confers increased stress resistance in brain and other tissues. To better understand the role of neuronal insulin-like growth factor signaling in neuroprotection, we inactivated insulin-like growth factor type-1-receptor in forebrain neurons using conditional Cre-LoxP-mediated gene targeting. We found that brain structure and function, including memory performance, were preserved in insulin-like growth factor receptor mutants, and that certain characteristics improved, notably synaptic transmission in hippocampal neurons. To reveal stress-related roles of insulin-like growth factor signaling, we challenged the brain using a stroke-like insult. Importantly, when charged with hypoxia-ischemia, mutant brains were broadly protected from cell damage, neuroinflammation and cerebral edema. We also found that in mice with insulin-like growth factor receptor knockout specifically in forebrain neurons, a substantial systemic upregulation of growth hormone and insulin-like growth factor-I occurred, which was associated with significant somatic overgrowth. Collectively, we found strong evidence that blocking neuronal insulin-like growth factor signaling increases peripheral somatotropic tone and simultaneously protects the brain against hypoxic-ischemic injury, findings that may contribute to developing new therapeutic concepts preventing the disabling consequences of stroke.


Subject(s)
Gene Deletion , Growth Hormone/metabolism , Neuroprotection , Prosencephalon/pathology , Receptor, IGF Type 1/genetics , Stroke/genetics , Stroke/pathology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Prosencephalon/metabolism , Stroke/metabolism , Up-Regulation
6.
Brain ; 139(Pt 4): 1237-51, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26912648

ABSTRACT

Recent studies highlight the implication of innate and adaptive immunity in the pathophysiology of Alzheimer's disease, and foster immunotherapy as a promising strategy for its treatment. Vaccines targeting amyloid-ß peptide provided encouraging results in mouse models, but severe side effects attributed to T cell responses in the first clinical trial AN1792 underlined the need for better understanding adaptive immunity in Alzheimer's disease. We previously showed that regulatory T cells critically control amyloid-ß-specific CD4(+) T cell responses in both physiological and pathological settings. Here, we analysed the impact of regulatory T cells on spontaneous disease progression in a murine model of Alzheimer's disease. Early transient depletion of regulatory T cells accelerated the onset of cognitive deficits in APPPS1 mice, without altering amyloid-ß deposition. Earlier cognitive impairment correlated with reduced recruitment of microglia towards amyloid deposits and altered disease-related gene expression profile. Conversely, amplification of regulatory T cells through peripheral low-dose IL-2 treatment increased numbers of plaque-associated microglia, and restored cognitive functions in APPPS1 mice. These data suggest that regulatory T cells play a beneficial role in the pathophysiology of Alzheimer's disease, by slowing disease progression and modulating microglial response to amyloid-ß deposition. Our study highlights the therapeutic potential of repurposed IL-2 for innovative immunotherapy based on modulation of regulatory T cells in Alzheimer's disease.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/pathology , Disease Progression , T-Lymphocytes, Regulatory/physiology , Alzheimer Disease/prevention & control , Amyloid beta-Protein Precursor , Animals , Brain/immunology , Brain/pathology , Humans , Interleukin-2/pharmacology , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1 , T-Lymphocytes, Regulatory/drug effects
7.
J Neurosci ; 35(34): 12063-79, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26311784

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disease characterized by the selective loss of spinal motor neurons due to the depletion of the survival of motor neuron (SMN) protein. No therapy is currently available for SMA, which represents the leading genetic cause of death in childhood. In the present study, we report that insulin-like growth factor-1 receptor (Igf-1r) gene expression is enhanced in the spinal cords of SMA-like mice. The reduction of expression, either at the physiological (through physical exercise) or genetic level, resulted in the following: (1) a significant improvement in lifespan and motor behavior, (2) a significant motor neuron protection, and (3) an increase in SMN expression in spinal cord and skeletal muscles through both transcriptional and posttranscriptional mechanisms. Furthermore, we have found that reducing IGF-1R expression is sufficient to restore intracellular signaling pathway activation profile lying downstream of IGF-1R, resulting in both the powerful activation of the neuroprotective AKT/CREB pathway and the inhibition of the ERK and JAK pathways. Therefore, reducing rather than enhancing the IGF-1 pathway could constitute a useful strategy to limit neurodegeneration in SMA. SIGNIFICANCE STATEMENT: Recent evidence of IGF-1 axis alteration in spinal muscular atrophy (SMA), a very severe neurodegenerative disease affecting specifically the motor neurons, have triggered a renewed interest in insulin-like growth factor-1 (IGF-1) pathway activation as a potential therapeutic approach for motor neuron diseases. The present study challenges this point of view and brings the alternative hypothesis that reducing rather than enhancing the IGF-1 signaling pathway exerts a neuroprotective effect in SMA. Furthermore, the present data substantiate a newly emerging concept that the modulation of IGF-1 receptor expression is a key event selectively determining the activation level of intracellular pathways that lie downstream of the receptor. This aspect should be considered when designing IGF-1-based treatments for neurodegenerative diseases.


Subject(s)
Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/prevention & control , Receptor, IGF Type 1/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscular Atrophy, Spinal/genetics , Receptor, IGF Type 1/genetics
8.
J Neurosci ; 35(33): 11500-13, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26290229

ABSTRACT

Alzheimer's disease (AD) is a frequent and irreversible age-related neurodegeneration without efficient treatment. Experimental AD in mice responds positively to decreased insulin-like growth factor I (IGF-I) signaling, a pathway also implicated in aging. Here we aimed to protect the aging brain from devastating amyloid pathology by making specifically adult neurons resistant to IGF signaling. To achieve that, we knocked out neuronal IGF-1R during adulthood in APP/PS1 mice. We found that mutants exhibited improved spatial memory and reduced anxiety. Mutant brains displayed fewer amyloid plaques, less amyloid-ß (Aß), and diminished neuroinflammation. Surprisingly, adult neurons undergoing IGF-1R knock-out reduced their apical soma and developed leaner dendrites, indicative of remarkable structural plasticity entailing condensed forebrain neuroarchitecture. Neurons lacking IGF-1R in AD showed less accumulation of Aß-containing autophagic vacuoles. At the same time, plasma Aß levels were increased. Our data indicate that neuronal IGF-1R ablation, via preserved autophagic compartment and enhanced systemic elimination, offers lifelong protection from AD pathology by clearing toxic Aß. Neuronal IGF-1R, and possibly other cell size-controlling pathways are promising targets for AD treatment. SIGNIFICANCE STATEMENT: We found compelling evidence in vivo that Alzheimer's disease (AD) progression is significantly delayed when insulin-like growth factor (IGF) signaling is blocked in adult neurons. To show that, we built a novel mouse model, combining inducible neuron-specific IGF-1R knock-out with AD transgenics. Analysis of the experimental AD phenotype revealed less abundant amyloid-ß (Aß) peptides, fewer plaques, and diminished neuroinflammation in mutants with inactivated IGF signaling, together with clearly preserved behavioral and memory performances. We present for the first time evidence that IGF signaling has profound effects on neuronal proteostasis and maintenance of cell morphology in vivo. Our results indicate in a model highly pertinent to translational research that neuronal IGF resistance may represent a pathophysiologically relevant mechanism of the brain for preventing Aß accumulation.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Insulin-Like Growth Factor I/metabolism , Neurons/metabolism , Neurons/pathology , Animals , Behavior, Animal , Cell Size , Cells, Cultured , Down-Regulation , Female , Male , Maze Learning , Metabolic Clearance Rate , Mice , Mice, Knockout , Mice, Transgenic , Prosencephalon/metabolism , Prosencephalon/pathology , Receptor, IGF Type 1/genetics , Signal Transduction
9.
Aging Cell ; 14(5): 847-56, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26219530

ABSTRACT

Downregulation of insulin-like growth factor (IGF) pathways prolongs lifespan in various species, including mammals. Still, the cellular mechanisms by which IGF signaling controls the aging trajectory of individual organs are largely unknown. Here, we asked whether suppression of IGF-I receptor (IGF-1R) in adult stem cells preserves long-term cell replacement, and whether this may prevent age-related functional decline in a regenerating tissue. Using neurogenesis as a paradigm, we showed that conditional knockout of IGF-1R specifically in adult neural stem cells (NSC) maintained youthful characteristics of olfactory bulb neurogenesis within an aging brain. We found that blocking IGF-I signaling in neural precursors increased cumulative neuroblast production and enhanced neuronal integration into the olfactory bulb. This in turn resulted in neuro-anatomical changes that improved olfactory function. Interestingly, mutants also displayed long-term alterations in energy metabolism, possibly related to IGF-1R deletion in NSCs throughout lifespan. We explored Akt and ERK signaling cascades and revealed differential regulation downstream of IGF-1R, with Akt phosphorylation preferentially decreased in IGF-1R(-/-) NSCs within the niche, and ERK pathway downregulated in differentiated neurons of the OB. These challenging experimental results were sustained by data from mathematical modeling, predicting that diminished stimulation of growth is indeed optimal for tissue aging. Thus, inhibiting growth and longevity gene IGF-1R in adult NSCs induced a gain-of-function phenotype during aging, marked by optimized management of cell renewal, and enhanced olfactory sensory function.


Subject(s)
Aging/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Olfactory Bulb/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Animals , Mice , Mice, Knockout , Receptor, IGF Type 1/deficiency , Receptor, IGF Type 1/genetics
10.
J Neurochem ; 124(1): 59-68, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23083210

ABSTRACT

Several epidemiological and preclinical studies suggest that non-steroidal anti-inflammatory drugs (NSAIDs), which inhibit cyclooxygenase (COX), reduce the risk of Alzheimer's disease (AD) and can lower ß-amyloid (Aß) production and inhibit neuroinflammation. However, follow-up clinical trials, mostly using selective cyclooxygenase (COX)-2 inhibitors, failed to show any beneficial effect in AD patients with mild to severe cognitive deficits. Recent data indicated that COX-1, classically viewed as the homeostatic isoform, is localized in microglia and is actively involved in brain injury induced by pro-inflammatory stimuli including Aß, lipopolysaccharide, and interleukins. We hypothesized that neuroinflammation is critical for disease progression and selective COX-1 inhibition, rather than COX-2 inhibition, can reduce neuroinflammation and AD pathology. Here, we show that treatment of 20-month-old triple transgenic AD (3 × Tg-AD) mice with the COX-1 selective inhibitor SC-560 improved spatial learning and memory, and reduced amyloid deposits and tau hyperphosphorylation. SC-560 also reduced glial activation and brain expression of inflammatory markers in 3 × Tg-AD mice, and switched the activated microglia phenotype promoting their phagocytic ability. The present findings are the first to demonstrate that selective COX-1 inhibition reduces neuroinflammation, neuropathology, and improves cognitive function in 3 × Tg-AD mice. Thus, selective COX-1 inhibition should be further investigated as a potential therapeutic approach for AD.


Subject(s)
Alzheimer Disease/complications , Amyloidogenic Proteins/metabolism , Cyclooxygenase Inhibitors/therapeutic use , Memory Disorders/drug therapy , Memory Disorders/etiology , Pyrazoles/therapeutic use , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Mutation/genetics , Phagocytes/drug effects , Phosphorylation/drug effects , Presenilin-1/genetics , tau Proteins/genetics
11.
J Neurochem ; 120(2): 292-301, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22050439

ABSTRACT

Like macrophages, microglia are functionally polarized into different phenotypic activation states, referred as classical and alternative. The balance of the two phenotypes may be critical to ensure proper brain homeostasis, and may be altered in brain pathological states, such as Alzheimer's disease. We investigated the role of NADPH oxidase in microglial activation state using p47(phox) and gp91(phox) -deficient mice as well as apocynin, a NADPH oxidase inhibitor during neuroinflammation induced by an intracerebroventricular injection of LPS or Aß1₋42. We showed that NADPH oxidase plays a critical role in the modulation of microglial phenotype and subsequent inflammatory response. We demonstrated that inhibition of NADPH oxidase or gene deletion of its functional p47(phox) subunit switched microglial activation from a classical to an alternative state in response to an inflammatory challenge. Moreover, we showed a shift in redox state towards an oxidized milieu and that subpopulations of microglia retain their detrimental phenotype in Alzheimer's disease brains. Microglia can change their activation phenotype depending on NADPH oxidase-dependent redox state of microenvironment. Inhibition of NADPH oxidase represents a promising neuroprotective approach to reduce oxidative stress and modulate microglial phenotype towards an alternative state.


Subject(s)
Encephalitis/metabolism , Encephalitis/pathology , Frontal Lobe/enzymology , Gene Expression Regulation, Enzymologic/physiology , Microglia/physiology , NADPH Oxidases/metabolism , ATPases Associated with Diverse Cellular Activities , Acetophenones/therapeutic use , Aged , Alzheimer Disease/pathology , Animals , CD11b Antigen/genetics , CD11b Antigen/metabolism , DNA Helicases/deficiency , Encephalitis/chemically induced , Encephalitis/drug therapy , Enzyme Inhibitors/therapeutic use , Enzyme-Linked Immunosorbent Assay , Female , Frontal Lobe/pathology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Injections, Intraventricular , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Postmortem Changes , RNA, Messenger/metabolism , Receptors, Immunologic/deficiency
12.
Biochimie ; 93(1): 46-51, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20868723

ABSTRACT

Neuroinflammation has been implicated in the pathogenesis or the progression of a variety of acute and chronic neurological and neurodegenerative disorders, including Alzheimer's disease. Prostaglandin H synthases or cyclooxygenases (COX -1 and COX-2) play a central role in the inflammatory cascade by converting arachidonic acid into bioactive prostanoids. In this review, we highlighted recent experimental data that challenge the classical view that the inducible isoform COX-2 is the most appropriate target to treat neuroinflammation. First, we discuss data showing that COX-2 activity is linked to anti-inflammatory and neuroprotective actions and is involved in the generation of novel lipid mediators with pro-resolution properties. Then, we review recent data demonstrating that COX-1, classically viewed as the homeostatic isoform, is actively involved in brain injury induced by pro-inflammatory stimuli including Aß, lipopolysaccharide, IL-1ß, and TNF-α. Overall, we suggest revisiting the traditional views on the roles of each COX during neuroinflammation and we propose COX-1 inhibition as a viable therapeutic approach to treat CNS diseases with a marked inflammatory component.


Subject(s)
Arachidonic Acid/metabolism , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase Inhibitors , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Prostaglandins/metabolism , Animals , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Humans , Mice , Neurodegenerative Diseases/physiopathology , Rats
13.
J Neuroinflammation ; 7: 57, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20920207

ABSTRACT

BACKGROUND: Passive immunization with antibodies directed to Aß decreases brain Aß/amyloid burden and preserves memory in transgenic mouse models of Alzheimer's disease (AD). This therapeutic strategy is under intense scrutiny in clinical studies, but its application is limited by neuroinflammatory side effects (autoimmune encephalitis and vasogenic edema). METHODS: We intravenously administered the monoclonal Aß protofibril antibody PFA1 to aged (22 month) male and female 3 × tg AD mice with intermediate or advanced AD-like neuropathologies, respectively, and measured brain and serum Aß and CNS cytokine levels. We also examined 17 month old 3 × tg AD female mice with intermediate pathology to determine the effect of amyloid burden on responses to passive immunization. RESULTS: The 22 month old male mice immunized with PFA1 had decreased brain Aß, increased serum Aß, and no change in CNS cytokine levels. In contrast, 22 month old immunized female mice revealed no change in brain Aß, decreased serum Aß, and increased CNS cytokine levels. Identical experiments in younger (17 month old) female 3 × tg AD mice with intermediate AD-like neuropathologies revealed a trend towards decreased brain Aß and increased serum Aß accompanied by a decrease in CNS MCP-1. CONCLUSIONS: These data suggest that passive immunization with PFA1 in 3 × tg AD mice with intermediate disease burden, regardless of sex, is effective in mediating potentially therapeutic effects such as lowering brain Aß. In contrast, passive immunization of mice with a more advanced amyloid burden may result in potentially adverse effects (encephalitis and vasogenic edema) mediated by certain proinflammatory cytokines.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Vaccines/therapeutic use , Amyloid beta-Peptides/metabolism , Brain/pathology , Immunization, Passive , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/immunology , Animals , Blotting, Western , Brain/immunology , Brain/metabolism , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Male , Mice , Mice, Transgenic , tau Proteins/immunology , tau Proteins/metabolism
14.
Neurosci Lett ; 478(3): 113-8, 2010 Jul 12.
Article in English | MEDLINE | ID: mdl-20451580

ABSTRACT

Neuroinflammation is a critical component in the progression of several neurological and neurodegenerative diseases and cyclooxygenases (COX)-1 and -2 are key regulators of innate immune responses. We recently demonstrated that COX-1 deletion attenuates, whereas COX-2 deletion enhances, the neuroinflammatory response, blood-brain barrier permeability and leukocyte recruitment during lipopolysaccharide (LPS)-induced innate immune activation. Here, we used transgenic mice, which overexpressed human COX-2 via neuron-specific Thy-1 promoter (TgCOX-2), causing elevated prostaglandins (PGs) levels. We tested whether neuronal COX-2 overexpression affects the glial response to a single intracerebroventricular injection of LPS, which produces a robust neuroinflammatory reaction. Relative to non-transgenic controls (NTg), 7 month-old TgCOX-2 did not show any basal neuroinflammation, as assessed by gene expression of markers of inflammation and oxidative stress, neuronal damage, as assessed by Fluoro-JadeB staining, or systemic inflammation, as assessed by plasma levels of IL-1beta and corticosterone. Twenty-four hours after LPS injection, all mice showed increased microglial activation, as indicated by Iba1 immunostaining, neuronal damage, mRNA expression of cytokines (TNF-alpha, IL-6), reactive oxygen expressing enzymes (iNOS and NADPH oxidase subunits), endogenous COX-2, cPLA(2) and mPGES-1, and hippocampal and cortical IL-1beta levels. However, the increases were similar in TgCOX-2 and NTg. In NTg, LPS increased brain PGE(2) to the levels observed in TgCOX-2. These results suggest that PGs derived from neuronal COX-2 do not play a role in the neuroinflammatory response to acute activation of brain innate immunity. This is likely due to the direct effect of LPS on glial rather than neuronal cells.


Subject(s)
Brain/immunology , Brain/metabolism , Cyclooxygenase 2/metabolism , Encephalitis/metabolism , Neurons/metabolism , Prostaglandins/metabolism , Animals , Brain/pathology , Corticosterone/blood , Cyclooxygenase 2/genetics , Encephalitis/chemically induced , Encephalitis/pathology , Humans , Interleukin-1beta/blood , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Neuroimmunomodulation/physiology , Neurons/pathology , Oxidative Stress/physiology , Prostaglandins E/metabolism , RNA, Messenger/metabolism
15.
J Cereb Blood Flow Metab ; 30(2): 370-80, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19844242

ABSTRACT

Cyclooxygenases (COX) -1 and -2 are key regulators of innate immune responses. We recently demonstrated that the expression of proinflammatory cytokines and chemokines is reduced in COX-1 null ((-/-)), and increased in COX-2(-/-) mice compared with their respective wild type controls during lipopolysaccharide (LPS)-induced innate immune activation. As chemokines are involved in leukocyte recruitment into the inflamed brain, we hypothesized that COX-1 and COX-2 deletion will differentially modulate blood-brain barrier (BBB) permeability in response to LPS. In the present study, using quantitative magnetic resonance imaging, we found that LPS-induced BBB disruption was exacerbated in COX-2(-/-) versus COX-2(+/+) mice. In the hippocampus and cortex of LPS-treated mice, matrix metalloproteinase (MMP)-3 activity was significantly decreased in COX-1(-/-) mice, whereas in COX-2(-/-) mice the activity of both MMP-9 and MMP-3, known to mediate BBB breakdown, was increased. Brain mRNA expression of the leukocyte attracting chemokine Cxcl10, the intercellular interaction molecule Icam-1, the pan-leukocyte marker Cd45 was increased in COX-2(-/-) versus COX-2(+/+) mice, whereas Cxcl10 and Cd45 mRNA expression was decreased in COX-1(-/-) versus COX-1(+/+) mice after LPS. Altogether, these results indicate that COX-2 activity modulates MMP-9 and-3 activities and is necessary to maintain BBB integrity during toll-like receptor 4-dependent innate immune activation.


Subject(s)
Blood-Brain Barrier/enzymology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 9/metabolism , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Capillary Permeability , Fluorescence Resonance Energy Transfer , Gene Expression/drug effects , Hippocampus/enzymology , Hippocampus/immunology , Hippocampus/pathology , Inflammation/enzymology , Inflammation/immunology , Injections, Intraventricular , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Magnetic Resonance Imaging , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
16.
Trends Pharmacol Sci ; 30(4): 174-81, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19269697

ABSTRACT

Cyclooxygenases (COX-1 and COX-2) are key enzymes in the conversion of arachidonic acid to prostaglandins and other lipid mediators. Because it can be induced by inflammatory stimuli, COX-2 has been classically considered as the most appropriate target for anti-inflammatory drugs. However, recent data indicate that COX-2 can mediate neuroprotection and that COX-1 is a major player in the neuroinflammatory process. We discuss the specific contributions of COX-1 and COX-2 in various neurodegenerative diseases and in models of neuroinflammation. We suggest that, owing to its predominant localization in microglia, COX-1 might be the major player in neuroinflammation, whereas COX-2, which is localized in neurons, might have a major role in models in which the neurons are directly challenged. Overall, the benefit of using COX-2 inhibitors should be carefully evaluated and COX-1 preferential inhibitors should be further investigated as a potential therapeutic approach in neurodegenerative diseases with an inflammatory component.


Subject(s)
Cyclooxygenase 1/physiology , Cyclooxygenase 2/physiology , Neuritis/pathology , Animals , Cyclooxygenase 2 Inhibitors/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Humans , Neuritis/drug therapy , Neurodegenerative Diseases/drug therapy
17.
J Neuroinflammation ; 5: 17, 2008 May 19.
Article in English | MEDLINE | ID: mdl-18489773

ABSTRACT

BACKGROUND: Cyclooxygenases (COX) -1 and -2 are key mediators of the inflammatory response in the central nervous system. Since COX-2 is inducible by inflammatory stimuli, it has been traditionally considered as the most appropriate target for anti-inflammatory drugs. However, the specific roles of COX-1 and COX-2 in modulating a neuroinflammatory response are unclear. Recently, we demonstrated that COX-1 deficient mice show decreased neuroinflammatory response and neuronal damage in response to lipopolysaccharide (LPS). METHODS: In this study, we investigated the role of COX-2 in the neuroinflammatory response to intracerebroventricular-injected LPS (5 mug), a model of direct activation of innate immunity, using COX-2 deficient (COX-2-/-) and wild type (COX-2+/+) mice, as well as COX-2+/+ mice pretreated for 6 weeks with celecoxib, a COX-2 selective inhibitor. RESULTS: Twenty-four hours after LPS injection, COX-2-/- mice showed increased neuronal damage, glial cell activation, mRNA and protein expression of markers of inflammation and oxidative stress, such as cytokines, chemokines, iNOS and NADPH oxidase. Brain protein levels of IL-1beta, NADPH oxidase subunit p67phox, and phosphorylated-signal transducer and activator of transcription 3 (STAT3) were higher in COX-2-/- and in celecoxib-treated mice, compared to COX-2+/+ mice. The increased neuroinflammatory response in COX-2-/- mice was likely mediated by the upregulation of STAT3 and suppressor of cytokine signaling 3 (SOCS3). CONCLUSION: These results show that inhibiting COX-2 activity can exacerbate the inflammatory response to LPS, possibly by increasing glial cells activation and upregulating the STAT3 and SOCS3 pathways in the brain.


Subject(s)
Cyclooxygenase 2 Inhibitors/toxicity , Cyclooxygenase 2/physiology , Encephalitis/enzymology , Lipopolysaccharides/toxicity , Nerve Tissue Proteins/biosynthesis , Animals , Celecoxib , Cyclooxygenase 1/physiology , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Cytokines/biosynthesis , Cytokines/genetics , Encephalitis/chemically induced , Encephalitis/immunology , Gene Expression Profiling , Lipopolysaccharides/administration & dosage , Male , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/etiology , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , Phosphorylation , Protein Processing, Post-Translational , Pyrazoles/toxicity , Reactive Oxygen Species , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Sulfonamides/toxicity , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
18.
Brain Res Bull ; 73(1-3): 108-13, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17499644

ABSTRACT

Brain aging is associated with inflammatory changes. However, data on how the brain arachidonic acid (AA) metabolism is altered as a function of age are limited and discrepant. AA is released from membrane phospholipids by phospholipase A(2) (PLA(2)) and then further metabolized to bioactive prostaglandins and thromboxanes by cyclooxygenases (COX)-1 and -2. We examined the phospholipase A(2) (PLA(2))/COX-mediated AA metabolic pathway in the hippocampus and cerebral cortex of 4-, 12-, 24- and 30-month-old rats. A two-fold increase in brain thromboxane B(2) level in 24 and 30 months was accompanied by increased hippocampal COX-1 mRNA levels at 12, 24, and 30 months. COX-2 mRNA expression was significantly decreased only at 30 months. Hippocampal Ca(2+)-independent iPLA(2) mRNA levels were decreased at 24 and 30 months without any change in Ca(2+)-dependent PLA(2) expression. In the cerebral cortex, mRNA levels of COX and PLA(2) were not significantly changed. The specific changes in the AA cascade observed in the hippocampus may alter phospholipids homeostasis and possibly increase the susceptibility of the aging brain to neuroinflammation.


Subject(s)
Aging/physiology , Calcium/physiology , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 1/genetics , Gene Expression Regulation, Enzymologic/drug effects , Hippocampus/enzymology , Hippocampus/growth & development , Phospholipases A/biosynthesis , Phospholipases A/genetics , Animals , Brain Chemistry/drug effects , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Dinoprostone/biosynthesis , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Male , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Thromboxane B2/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...