Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
J Neuroinflammation ; 21(1): 93, 2024 Apr 15.
Article En | MEDLINE | ID: mdl-38622654

The neuroinflammatory process in synucleinopathies of the aging population such as Parkinson's disease (PD) and dementia with Lewy bodies (DLB) involves microglial activation as well as infiltration of the CNS by T cells and natural killer T cells (NKTs). To evaluate the potential of targeting NKT cells to modulate neuroinflammation, we treated α-syn transgenic (tg) mice (e.g.: Thy1 promoter line 61) with an antibody against CD1d, which is a glycoprotein expressed in antigen presenting cells (APCs). CD1d-presented lipid antigens activate NKT cells through the interaction with T cell receptor in NKTs, resulting in the production of cytokines. Thus, we hypothesized that blocking the APC-NKT interaction with an anti-CD1d antibody might reduce neuroinflammation and neurodegeneration in models of DLB/PD. Treatment with the anti-CD1d antibody did not have effects on CD3 (T cells), slightly decreased CD4 and increased CD8 lymphocytes in the mice. Moreover, double labeling studies showed that compared to control (IgG) treated α-syn tg mice, treatment with anti-CD1d decreased numbers of CD3/interferon γ (IFN γ)-positive cells, consistent with NKTs. Further double labeling studies showed that CD1d-positive cells co-localized with the astrocytes marker GFAP and that anti-CD1d antibody reduced this effect. While in control α-syn tg mice CD3 positive cells were near astrocytes, this was modified by the treatment with the CD1d antibody. By qPCR, levels of IFN γ, CCL4, and interleukin-6 were increased in the IgG treated α-syn tg mice. Treatment with CD1d antibody blunted this cytokine response that was associated with reduced astrocytosis and microgliosis in the CNS of the α-syn tg mice treated with CD1d antibody. Flow cytometric analysis of immune cells in α-syn tg mice revealed that CD1d-tet + T cells were also increased in the spleen of α-syn tg mice, which treatment with the CD1d antibody reduced. Reduced neuroinflammation in the anti-CD1d-treated α-syn tg mice was associated with amelioration of neurodegenerative pathology. These results suggest that reducing infiltration of NKT cells with an antibody against CD1d might be a potential therapeutical approach for DLB/PD.


Parkinson Disease , alpha-Synuclein , Mice , Animals , alpha-Synuclein/genetics , Lewy Bodies/pathology , Neuroinflammatory Diseases , Parkinson Disease/pathology , Mice, Transgenic , Immunotherapy/methods , Cytokines , Immunoglobulin G
3.
Transfusion ; 63(12): 2265-2272, 2023 12.
Article En | MEDLINE | ID: mdl-37850496

BACKGROUND: The burden of transfusion-transmitted infections among blood recipients remains low due to extensive pre- and post-donation screening. However, the military has the unique challenge of providing blood in austere environments with limited testing capabilities. This study evaluates the infectious etiologies of deferred blood donors at a large military blood donation center. METHODS: All blood donors at the Armed Service Blood Bank Center, San Antonio, between 2017 and 2022 with positive post-donation screening for hepatitis C (HCV), hepatitis B (HBV), human immunodeficiency virus (HIV), human T-lymphotropic virus (HTLV-I/II), Zika (2018-2021), West Nile virus, Trypanosoma cruzi, Treponema pallidum, or Babesia microti (2020-2022) were evaluated. Donors were deferred based on Food and Drug Administration (FDA) guidance. RESULTS: Two-hundred and thirteen (213) donors met FDA criteria for deferral. T. pallidum (n = 45, 50.3 per 100,000), HCV (n = 34, 38.0 per 100,000), and HBV (n = 19, 21.2 per 100,000) were the most common pathogens among those with both positive screening and confirmatory testing. The majority of HIV (95%), Chagas (78%), HTLV-I/II (50%) deferrals were due to indeterminate confirmatory tests following initial positive screens. The majority of deferrals for HBV were for a second occurrence of a positive screen despite negative confirmatory testing. CONCLUSION: The rates of post-donation deferral for transfusion-transmissible infections were low in this military cohort. Our findings suggest that donor testing in deployed service members should focus on HBV, HCV, and T. pallidum and highlight the need for better diagnostics for HIV, Chagas, and HTLV-I/II.


HIV Infections , Hepatitis B , Hepatitis C , Military Personnel , Zika Virus Infection , Zika Virus , Humans , Blood Donation , Hepatitis B/epidemiology , Hepatitis C/epidemiology , Hepacivirus , HIV , Blood Donors , HIV Infections/epidemiology
4.
Sci Transl Med ; 15(695): eabq6089, 2023 05 10.
Article En | MEDLINE | ID: mdl-37163617

Alterations in the p38 mitogen-activated protein kinases (MAPKs) play an important role in the pathogenesis of dementia with Lewy bodies (DLB) and Parkinson's disease (PD). Activation of the p38α MAPK isoform and mislocalization of the p38γ MAPK isoform are associated with neuroinflammation and synaptic degeneration in DLB and PD. Therefore, we hypothesized that p38α might be associated with neuronal p38γ distribution and synaptic dysfunction in these diseases. To test this hypothesis, we treated in vitro cellular and in vivo mouse models of DLB/PD with SKF-86002, a compound that attenuates inflammation by inhibiting p38α/ß, and then investigated the effects of this compound on p38γ and neurodegenerative pathology. We found that inhibition of p38α reduced neuroinflammation and ameliorated synaptic, neurodegenerative, and motor behavioral deficits in transgenic mice overexpressing human α-synuclein. Moreover, treatment with SKF-86002 promoted the redistribution of p38γ to synapses and reduced the accumulation of α-synuclein in mice overexpressing human α-synuclein. Supporting the potential value of targeting p38 in DLB/PD, we found that SKF-86002 promoted the redistribution of p38γ in neurons differentiated from iPS cells derived from patients with familial PD (carrying the A53T α-synuclein mutation) and healthy controls. Treatment with SKF-86002 ameliorated α-synuclein-induced neurodegeneration in these neurons only when microglia were pretreated with this compound. However, direct treatment of neurons with SKF-86002 did not affect α-synuclein-induced neurotoxicity, suggesting that SKF-86002 treatment inhibits α-synuclein-induced neurotoxicity mediated by microglia. These findings provide a mechanistic connection between p38α and p38γ as well as a rationale for targeting this pathway in DLB/PD.


Mitogen-Activated Protein Kinase 14 , Parkinson Disease , Humans , Mice , Animals , Parkinson Disease/drug therapy , Parkinson Disease/pathology , alpha-Synuclein/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Neuroinflammatory Diseases , Neurons/metabolism , Mice, Transgenic
5.
Mol Neurodegener ; 17(1): 60, 2022 09 05.
Article En | MEDLINE | ID: mdl-36064424

BACKGROUND: Although ɑ-synuclein (ɑ-syn) spreading in age-related neurodegenerative diseases such as Parkinson's disease (PD) and Dementia with Lewy bodies (DLB) has been extensively investigated, the role of aging in the manifestation of disease remains unclear. METHODS: We explored the role of aging and inflammation in the pathogenesis of synucleinopathies in a mouse model of DLB/PD initiated by intrastriatal injection of ɑ-syn preformed fibrils (pff). RESULTS: We found that aged mice showed more extensive accumulation of ɑ-syn in selected brain regions and behavioral deficits that were associated with greater infiltration of T cells and microgliosis. Microglial inflammatory gene expression induced by ɑ-syn-pff injection in young mice had hallmarks of aged microglia, indicating that enhanced age-associated pathologies may result from inflammatory synergy between aging and the effects of ɑ-syn aggregation. Based on the transcriptomics analysis projected from Ingenuity Pathway Analysis, we found a network that included colony stimulating factor 2 (CSF2), LPS related genes, TNFɑ and poly rl:rC-RNA as common regulators. CONCLUSIONS: We propose that aging related inflammation (eg: CSF2) influences outcomes of pathological spreading of ɑ-syn and suggest that targeting neuro-immune responses might be important in developing treatments for DLB/PD.


Parkinson Disease , Synucleinopathies , Animals , Brain/metabolism , Disease Models, Animal , Inflammation/metabolism , Mice , Parkinson Disease/metabolism , alpha-Synuclein/metabolism
6.
Article En | MEDLINE | ID: mdl-36011504

This study aimed to verify the relationship between handgrip strength and oral health using data from the Korea National Health and Nutrition Examination Survey, representing Korean adults. Data from the seventh survey (2016−2018) conducted by the Korea Centers for Disease Control and Prevention were utilized, and 10,607 final study participants were recruited according to the selection and exclusion criteria. A complex sample logistic regression analysis was performed to confirm the relationship between handgrip strength and dental caries according to the sex of the study participants. On analyzing the correlation in men, "C1 (DMFT: 11−32)," when compared to "C4 (DMFT: 0−3)," in Model 1 without adjustment for potential confounders, was 2.92 (95% confidence interval [CI]: 2.15−3.97) times more likely to be associated with lower handgrip strength, and a statistically significant result was detected (p < 0.001). Additionally, significant odds ratios (ORs) were confirmed for all adjusted models. In women, the ORs in Model 1 without adjustment for potential confounders were 1.41 times (95% CI: 1.14−1.75) and demonstrated a significant result; however, the results were not significant in all adjusted models 2−4. Resultantly, a significant association was detected between dental caries and handgrip strength in Korean adults.


Dental Caries , Hand Strength , Adult , Cross-Sectional Studies , Dental Caries/epidemiology , Female , Humans , Male , Nutrition Surveys , Republic of Korea/epidemiology
7.
Cells ; 10(4)2021 04 03.
Article En | MEDLINE | ID: mdl-33916798

Rheumatoid arthritis (RA) is a common autoimmune disease characterized by immune cell infiltration of the synovium, leading to the loss of cartilage, bone, and joint function. Although regulatory T (Treg) cells are thought to modulate the initiation and progression of RA, a consensus has yet to be reached regarding the function and composition of Treg cells in RA patients. To address these discrepancies, we analyzed not only the total Treg frequency but also that of Treg subpopulations in the peripheral blood of RA patients and healthy controls by flow cytometry. We found that the total Treg population was not significantly different between RA and control subjects. However, the effector Treg cell subgroup, defined as CD45RA-CD25hi, showed markedly decreased frequency in RA patients. In addition, the total Treg population from RA patients showed a significant decline in the expression of CD25. Both the naïve and effector Treg subgroups also showed marked reduction of CD25 expression in RA patients compared to controls. These data suggest that the decreased frequency of effector Treg cells and overall reduction of CD25 expression in Treg cells in the peripheral blood may be evidence of altered Treg homeostasis associated with RA pathogenesis.


Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , T-Lymphocytes, Regulatory/immunology , Adult , Case-Control Studies , Humans , Lymphocyte Count
8.
Exp Mol Med ; 53(2): 281-290, 2021 02.
Article En | MEDLINE | ID: mdl-33594256

Synucleinopathies are age-related neurological disorders characterized by the progressive deposition of α-synuclein (α-syn) aggregates and include Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Although cell-to-cell α-syn transmission is thought to play a key role in the spread of α-syn pathology, the detailed mechanism is still unknown. Neuroinflammation is another key pathological feature of synucleinopathies. Previous studies have identified several immune receptors that mediate neuroinflammation in synucleinopathies, such as Toll-like receptor 2 (TLR2). However, the species of α-syn aggregates varies from study to study, and how different α-syn aggregate species interact with innate immune receptors has yet to be addressed. Therefore, we investigated whether innate immune receptors can facilitate the uptake of different species of α-syn aggregates. Here, we examined whether stimulation of TLRs could modulate the cellular uptake and degradation of α-syn fibrils despite a lack of direct interaction. We observed that stimulation of TLR2 in vitro accelerated α-syn fibril uptake in neurons and glia while delaying the degradation of α-syn in neurons and astrocytes. Internalized α-syn was rapidly degraded in microglia regardless of whether TLR2 was stimulated. However, cellular α-syn uptake and degradation kinetics were not altered by TLR4 stimulation. In addition, upregulation of TLR2 expression in a synucleinopathy mouse model increased the density of Lewy-body-like inclusions and induced morphological changes in microglia. Together, these results suggest that cell type-specific modulation of TLR2 may be a multifaceted and promising therapeutic strategy for synucleinopathies; inhibition of neuronal and astroglial TLR2 decreases pathogenic α-syn transmission, but activation of microglial TLR2 enhances microglial extracellular α-syn clearance.


Brain/immunology , Brain/metabolism , Immunity, Innate , Receptors, Immunologic/agonists , Receptors, Immunologic/metabolism , alpha-Synuclein/metabolism , Animals , Astrocytes , Brain/pathology , Cell Line , Extracellular Space/metabolism , Humans , Immunity, Innate/genetics , Immunohistochemistry , Mice , Mice, Transgenic , Microglia/metabolism , Models, Animal , Models, Biological , Neurons/metabolism , Protease Inhibitors/pharmacology , Protein Binding , Protein Transport , Proteolysis , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , alpha-Synuclein/genetics
9.
Sci Transl Med ; 12(565)2020 10 14.
Article En | MEDLINE | ID: mdl-33055242

Synucleinopathies are neurodegenerative disorders characterized by abnormal α-synuclein deposition that include Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. The pathology of these conditions also includes neuronal loss and neuroinflammation. Neuron-released α-synuclein has been shown to induce neurotoxic, proinflammatory microglial responses through Toll-like receptor 2, but the molecular mechanisms involved are poorly understood. Here, we show that leucine-rich repeat kinase 2 (LRRK2) plays a critical role in the activation of microglia by extracellular α-synuclein. Exposure to α-synuclein was found to enhance LRRK2 phosphorylation and activity in mouse primary microglia. Furthermore, genetic and pharmacological inhibition of LRRK2 markedly diminished α-synuclein-mediated microglial neurotoxicity via lowering of tumor necrosis factor-α and interleukin-6 expression in mouse cultures. We determined that LRRK2 promoted a neuroinflammatory cascade by selectively phosphorylating and inducing nuclear translocation of the immune transcription factor nuclear factor of activated T cells, cytoplasmic 2 (NFATc2). NFATc2 activation was seen in patients with synucleinopathies and in a mouse model of synucleinopathy, where administration of an LRRK2 pharmacological inhibitor restored motor behavioral deficits. Our results suggest that modulation of LRRK2 and its downstream signaling mediator NFATc2 might be therapeutic targets for treating synucleinopathies.


Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Microglia , NFATC Transcription Factors/metabolism , Synucleinopathies , Animals , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice , Rodentia , Transcription Factors , alpha-Synuclein
10.
Nutrients ; 12(9)2020 Sep 09.
Article En | MEDLINE | ID: mdl-32916825

Despite the advancements in vaccination research and practices, influenza viruses remain a global health concern. Inducing a robust immune response by vaccination is especially challenging in the elderly, the immunocompromised, and persons with chronic illnesses. Polysaccharides derived from food may act as a safe and readily accessible means to boost the immune system during vaccination. In this study, we investigated whether crude polysaccharides derived from carrot pomace (CPP) could stimulate innate immune cell function and promote influenza vaccine immunogenicity. In bone marrow-derived dendritic cells (BMDCs), CPP increased the fraction of CD11c+MHCII+ cells and the expression of co-stimulatory molecules CD40 and CD80, indicative of enhanced maturation and activation. Functionally, CPP-treated BMDCs promoted inflammatory cytokine production in splenic lymphocytes. In a mouse model of immunosuppression induced by cyclophosphamide, animals given CPP before and after an influenza vaccine challenge showed increased frequencies of dendritic cells and natural killer cells in the spleen, in addition to the recovery of vaccine-specific antibody titers. Moreover, innate myeloid cells in CPP-fed mice showed evidence of phenotypic modification via markedly enhanced interleukin(IL)-12 and interferon(IFN)-γ production in response to lipopolysaccharide(LPS) stimulation ex vivo. Our findings suggest that the administration of carrot pomace polysaccharides can significantly enhance the efficacy of influenza vaccination.


Daucus carota/chemistry , Dendritic Cells/immunology , Immunogenicity, Vaccine/drug effects , Influenza Vaccines/immunology , Polysaccharides/pharmacology , Animals , Immunity, Innate/drug effects , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control
11.
J Neuroinflammation ; 17(1): 214, 2020 Jul 17.
Article En | MEDLINE | ID: mdl-32680537

BACKGROUND: α-Synuclein (α-syn) is a pre-synaptic protein which progressively accumulates in neuronal and non-neuronal cells in neurodegenerative diseases such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy. Recent evidence suggests that aberrant immune activation may be involved in neurodegeneration in PD/DLB. While previous studies have often focused on the microglial responses, less is known about the role of the peripheral immune system in these disorders. METHODS: To understand the involvement of the peripheral immune system in PD/DLB, we evaluated T cell populations in the brains of α-syn transgenic (tg) mice (e.g., Thy1 promoter line 61) and DLB patients. RESULTS: Immunohistochemical analysis showed perivascular and parenchymal infiltration by CD3+/CD4+ helper T cells, but not cytotoxic T cells (CD3+/CD8+) or B cells (CD20+), in the neocortex, hippocampus, and striatum of α-syn tg mice. CD3+ cells were found in close proximity to the processes of activated astroglia, particularly in areas of the brain with significant astrogliosis, microgliosis, and expression of pro-inflammatory cytokines. In addition, a subset of CD3+ cells co-expressed interferon γ. Flow cytometric analysis of immune cells in the brains of α-syn tg mice revealed that CD1d-tet+ T cells were also increased in the brains of α-syn tg mice suggestive of natural killer T cells. In post-mortem DLB brains, we similarly detected increased numbers of infiltrating CD3+/CD4+ T cells in close proximity with blood vessels. CONCLUSION: These results suggest that infiltrating adaptive immune cells play an important role in neuroinflammation and neurodegeneration in synucleinopathies and that modulating peripheral T cells may be a viable therapeutic strategy for PD/DLB.


Adaptive Immunity/physiology , Brain/metabolism , Lewy Body Disease/metabolism , T-Lymphocytes/metabolism , alpha-Synuclein/metabolism , Aged , Aged, 80 and over , Animals , Brain/immunology , Brain/pathology , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lewy Body Disease/immunology , Lewy Body Disease/pathology , Male , Mice , Mice, Transgenic , T-Lymphocytes/immunology , T-Lymphocytes/pathology , alpha-Synuclein/immunology
12.
Neurotherapeutics ; 17(3): 935-954, 2020 07.
Article En | MEDLINE | ID: mdl-32347461

Neurological disorders such as Alzheimer's disease (AD), Lewy body dementia (LBD), frontotemporal dementia (FTD), and vascular dementia (VCID) have no disease-modifying treatments to date and now constitute a dementia crisis that affects 5 million in the USA and over 50 million worldwide. The most common pathological hallmark of these age-related neurodegenerative diseases is the accumulation of specific proteins, including amyloid beta (Aß), tau, α-synuclein (α-syn), TAR DNA-binding protein 43 (TDP43), and repeat-associated non-ATG (RAN) peptides, in the intra- and extracellular spaces of selected brain regions. Whereas it remains controversial whether these accumulations are pathogenic or merely a byproduct of disease, the majority of therapeutic research has focused on clearing protein aggregates. Immunotherapies have garnered particular attention for their ability to target specific protein strains and conformations as well as promote clearance. Immunotherapies can also be neuroprotective: by neutralizing extracellular protein aggregates, they reduce spread, synaptic damage, and neuroinflammation. This review will briefly examine the current state of research in immunotherapies against the 3 most commonly targeted proteins for age-related neurodegenerative disease: Aß, tau, and α-syn. The discussion will then turn to combinatorial strategies that enhance the effects of immunotherapy against aggregating protein, followed by new potential targets of immunotherapy such as aging-related processes.


Aging/drug effects , Drug Delivery Systems/methods , Immunologic Factors/administration & dosage , Immunotherapy/methods , Neurodegenerative Diseases/drug therapy , Aging/immunology , Aging/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/immunology , Brain/metabolism , Clinical Trials as Topic/methods , Drug Delivery Systems/trends , Gene-Environment Interaction , Humans , Immunologic Factors/immunology , Immunologic Factors/metabolism , Immunotherapy/trends , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/metabolism , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/immunology , alpha-Synuclein/metabolism , tau Proteins/antagonists & inhibitors , tau Proteins/immunology , tau Proteins/metabolism
13.
Front Neurosci ; 14: 286, 2020.
Article En | MEDLINE | ID: mdl-32296304

Progressive accumulation of the pre-synaptic protein α-synuclein (α-syn) has been strongly associated with the pathogenesis of neurodegenerative disorders of the aging population such as Alzheimer's disease (AD), Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy. While the precise mechanisms are not fully understood, alterations in kinase pathways including that of mitogen activated protein kinase (MAPK) p38 have been proposed to play a role. In AD, p38α activation has been linked to neuro-inflammation while alterations in p38γ have been associated with tau phosphorylation. Although p38 has been studied in AD, less is known about its role in DLB/PD and other α-synucleinopathies. For this purpose, we investigated the expression of the p38 family in brains from α-syn overexpressing transgenic mice (α-syn Tg: Line 61) and patients with DLB/PD. Immunohistochemical analysis revealed that in healthy human controls and non-Tg mice, p38α associated with neurons and astroglial cells and p38γ localized to pre-synaptic terminals. In DLB and α-syn Tg brains, however, p38α levels were increased in astroglial cells while p38γ immunostaining was redistributed from the synaptic terminals to the neuronal cell bodies. Double immunolabeling further showed that p38γ colocalized with α-syn aggregates in DLB patients, and immunoblot and qPCR analysis confirmed the increased levels of p38α and p38γ. α1-syntrophin, a synaptic target of p38γ, was present in the neuropil and some neuronal cell bodies in human controls and non-Tg mice. In DLB and and Tg mice, however, α1-syntrophin was decreased in the neuropil and instead colocalized with α-syn in intra-neuronal inclusions. In agreement with these findings, in vitro studies showed that α-syn co-immunoprecipitates with p38γ, but not p38α. These results suggest that α-syn might interfere with the p38γ pathway and play a role in the mechanisms of synaptic dysfunction in DLB/PD.

14.
Exp Neurobiol ; 28(5): 547-553, 2019 Oct 31.
Article En | MEDLINE | ID: mdl-31698547

Synucleinopathies are neurodegenerative disorders characterized by the progressive accumulation of α-synuclein (α-syn) in neurons and glia and include Parkinson's disease (PD) and dementia with Lewy bodies (DLB). In this review, we consolidate our key findings and recent studies concerning the role of Toll-like receptor 2 (TLR2), a pattern recognition innate immune receptor, in the pathogenesis of synucleinopathies. First, we address the pathological interaction of α-syn with microglial TLR2 and its neurotoxic inflammatory effects. Then, we show that neuronal TLR2 activation not only induces abnormal α-syn accumulation by impairing autophagy, but also modulates α-syn transmission. Finally, we demonstrate that administration of a TLR2 functional inhibitor improves the neuropathology and behavioral deficits of a synucleinopathy mouse model. Altogether, we present TLR2 modulation as a promising immunotherapy for synucleinopathies.

15.
Nat Neurosci ; 18(7): 1017-24, 2015 Jul.
Article En | MEDLINE | ID: mdl-26005851

Resilience to aversive events has a central role in determining whether stress leads to the development of depression. mGluR5 has been implicated in the pathophysiology of depression, but the effect of mGluR5 activity on stress resilience remains unexplored. We found that mGluR5(-/-) (also known as Grm5(-/-)) mice displayed more depression-like behaviors (for example, learned helplessness, social withdrawal and anhedonia) than control mice following exposure to various stressful stimuli. Lentiviral 'rescue' of mGluR5 in the nucleus accumbens (NAc) decreased these depression-like behaviors in mGluR5(-/-) mice. In the NAc, ΔFosB, whose induction promotes stress resilience, failed to be upregulated by stress in mGluR5(-/-) mice. Notably, targeted pharmacological activation of mGluR5 in the NAc increased ΔFosB expression. Our findings point to an essential role for mGluR5 in promoting stress resilience and suggest that a defect in mGluR5-mediated signaling in the NAc may represent an endophenotype for stress-induced depression.


Behavior, Animal/physiology , Depression/metabolism , Nucleus Accumbens/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptor, Metabotropic Glutamate 5/physiology , Resilience, Psychological , Signal Transduction/physiology , Stress, Psychological/metabolism , Animals , Depression/etiology , Disease Models, Animal , Endophenotypes , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Receptor, Metabotropic Glutamate 5/metabolism , Stress, Psychological/complications
16.
Metabolism ; 63(11): 1455-61, 2014 Nov.
Article En | MEDLINE | ID: mdl-25200186

OBJECTIVE: Estrogen plays an important role in the control of energy balance in the hypothalamus. Leptin-independent STAT3 activation (i.e., tyrosine(705)-phosphorylation of STAT3, pSTAT3) in the hypothalamus is hypothesized as the primary mechanism of the estrogen-induced anorexic response. However, the type of estrogen receptor that mediates this regulation is unknown. We investigated the role of the G protein-coupled receptor 30 (GPR30) in estradiol (E2)-induced STAT3 activation in the hypothalamus. MATERIALS/METHODS: Regulation of STAT3 activation by E2, G-1, a specific agonist of GPR30 and G-15, a specific antagonist of GPR30 was analyzed in vitro and in vivo. Effect of GPR30 activation on eating behavior was analyzed in vivo. RESULTS: E2 stimulated pSTAT3 in cells expressing GPR30, but not expressing estrogen receptor ERα and ERß. G-1 induced pSTAT3, and G-15 inhibited E2-induced pSTAT3 in primary cultures of hypothalamic neurons. A cerebroventricular injection of G-1 increased pSTAT3 in the arcuate nucleus of mice, which was associated with a decrease in food intake and body weight gain. CONCLUSIONS: These results suggest that GPR30 is the estrogen receptor that mediates the anorectic effect of estrogen through the STAT3 pathway in the hypothalamus.


Anorexia/physiopathology , Estrogens/physiology , Hypothalamus/physiopathology , Receptors, Estrogen/physiology , Receptors, G-Protein-Coupled/physiology , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Animals , Base Sequence , DNA Primers , HeLa Cells , Humans , Mice , Reverse Transcriptase Polymerase Chain Reaction
...