Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cell ; 184(24): 5902-5915.e17, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34752731

ABSTRACT

Increasing evidence indicates that the brain regulates peripheral immunity, yet whether and how the brain represents the state of the immune system remains unclear. Here, we show that the brain's insular cortex (InsCtx) stores immune-related information. Using activity-dependent cell labeling in mice (FosTRAP), we captured neuronal ensembles in the InsCtx that were active under two different inflammatory conditions (dextran sulfate sodium [DSS]-induced colitis and zymosan-induced peritonitis). Chemogenetic reactivation of these neuronal ensembles was sufficient to broadly retrieve the inflammatory state under which these neurons were captured. Thus, we show that the brain can store and retrieve specific immune responses, extending the classical concept of immunological memory to neuronal representations of inflammatory information.


Subject(s)
Immunity , Insular Cortex/physiology , Neurons/physiology , Animals , Colitis/chemically induced , Colitis/complications , Colitis/immunology , Colon/pathology , Dextran Sulfate , Female , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Peritoneum/pathology , Peritonitis/complications , Peritonitis/immunology , Peritonitis/pathology , Synapses/metabolism , Zymosan
3.
Mol Psychiatry ; 28(3): 1170-1181, 2023 03.
Article in English | MEDLINE | ID: mdl-36380234

ABSTRACT

Ample evidence implicate mitochondria in early brain development. However, to the best of our knowledge, there is only circumstantial data for mitochondria involvement in late brain development occurring through adolescence, a critical period in the pathogenesis of various psychiatric disorders, specifically schizophrenia. In schizophrenia, neurodevelopmental abnormalities and mitochondrial dysfunction has been repeatedly reported. Here we show a causal link between mitochondrial transplantation in adolescence and brain functioning in adulthood. We show that transplantation of allogenic healthy mitochondria into the medial prefrontal cortex of adolescent rats was beneficial in a rat model of schizophrenia, while detrimental in healthy control rats. Specifically, disparate initial changes in mitochondrial function and inflammatory response were associated with opposite long-lasting changes in proteome, neurotransmitter turnover, neuronal sprouting and behavior in adulthood. A similar inverse shift in mitochondrial function was also observed in human lymphoblastoid cells deived from schizophrenia patients and healthy subjects due to the interference of the transplanted mitochondria with their intrinsic mitochondrial state. This study provides fundamental insights into the essential role of adolescent mitochondrial homeostasis in the development of normal functioning adult brain. In addition, it supports a therapeutic potential for mitochondria manipulation in adolescence in disorders with neurodevelopmental and bioenergetic deficits, such as schizophrenia, yet emphasizes the need to monitor individuals' state including their mitochondrial function and immune response, prior to intervention.


Subject(s)
Schizophrenia , Adult , Rats , Humans , Animals , Adolescent , Mitochondria , Brain , Neurons , Disease Models, Animal
4.
J Neuroinflammation ; 20(1): 174, 2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37496076

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is the leading cause of dementia in the world. The pathology of AD is affiliated with the elevation of both tau (τ) and ß-amyloid (Aß) pathologies. Yet, the direct link between natural τ expression on glia cell activity and Aß remains unclear. While experiments in mouse models suggest that an increase in Aß exacerbates τ pathology when expressed under a neuronal promoter, brain pathology from AD patients suggests an appearance of τ pathology in regions without Aß. METHODS: Here, we aimed to assess the link between τ and Aß using a new mouse model that was generated by crossing a mouse model that expresses two human mutations of the human MAPT under a mouse Tau natural promoter with 5xFAD mice that express human mutated APP and PS1 in neurons. RESULTS: The new mouse model, called 5xFAD TAU, shows accelerated cognitive impairment at 2 months of age, increased number of Aß depositions at 4 months and neuritic plaques at 6 months of age. An expression of human mutated TAU in astrocytes leads to a dystrophic appearance and reduces their ability to engulf Aß, which leads to an increased brain Aß load. Astrocytes expressing mutated human TAU showed an impairment in the expression of vascular endothelial growth factor (VEGF) that has previously been suggested to play an important role in supporting neurons. CONCLUSIONS: Our results suggest the role of τ in exacerbating Aß pathology in addition to pointing out the potential role of astrocytes in disease progression. Further research of the crosstalk between τ and Aß in astrocytes may increase our understanding of the role glia cells have in the pathology of AD with the aim of identifying novel therapeutic interventions to an otherwise currently incurable disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Animals , Humans , Infant , Mice , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Astrocytes/metabolism , Brain/metabolism , Disease Models, Animal , Mice, Transgenic , tau Proteins/genetics , tau Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism
5.
Glia ; 67(7): 1359-1373, 2019 07.
Article in English | MEDLINE | ID: mdl-30882931

ABSTRACT

The cross-talk between blood proteins, immune cells, and brain function involves complex mechanisms. Plasma protein C1 inhibitor (C1INH) is an inhibitor of vascular inflammation that is induced by activation of the kallikrein-kinin system (KKS) and the complement system. Knockout of C1INH was previously correlated with peripheral vascular permeability via the bradykinin pathway, yet there was no evidence of its correlation with blood-brain barrier (BBB) integrity and brain function. In order to understand the effect of plasma C1INH on brain pathology via the vascular system, we knocked down circulating C1INH in wild-type (WT) mice using an antisense oligonucleotide (ASO), without affecting C1INH expression in peripheral immune cells or the brain, and examined brain pathology. Long-term elimination of endogenous C1INH in the plasma induced the activation of the KKS and peritoneal macrophages but did not activate the complement system. Bradykinin pathway proteins were elevated in the periphery and the brain, resulting in hypotension. BBB permeability, extravasation of plasma proteins into the brain parenchyma, activation of glial cells, and elevation of pro-inflammatory response mediators were detected. Furthermore, infiltrating innate immune cells were observed entering the brain through the lateral ventricle walls and the neurovascular unit. Mice showed normal locomotion function, yet cognition was impaired and depressive-like behavior was evident. In conclusion, our results highlight the important role of regulated plasma C1INH as it acts as a gatekeeper to the brain via the neurovascular system. Thus, manipulation of C1INH in neurovascular disorders might be therapeutically beneficial.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Capillary Permeability/physiology , Complement C1 Inhibitor Protein/metabolism , Locomotion/physiology , Neuroglia/metabolism , Animals , Brain/blood supply , Complement C1 Inhibitor Protein/genetics , Female , Gene Knockdown Techniques/methods , Inflammation/genetics , Inflammation/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
6.
Am J Hum Genet ; 99(2): 501-10, 2016 Aug 04.
Article in English | MEDLINE | ID: mdl-27453578

ABSTRACT

Cell division terminates with cytokinesis and cellular separation. Autosomal-recessive primary microcephaly (MCPH) is a neurodevelopmental disorder characterized by a reduction in brain and head size at birth in addition to non-progressive intellectual disability. MCPH is genetically heterogeneous, and 16 loci are known to be associated with loss-of-function mutations predominantly affecting centrosomal-associated proteins, but the multiple roles of centrosomes in cellular function has left questions about etiology. Here, we identified three families affected by homozygous missense mutations in CIT, encoding citron rho-interacting kinase (CIT), which has established roles in cytokinesis. All mutations caused substitution of conserved amino acid residues in the kinase domain and impaired kinase activity. Neural progenitors that were differentiated from induced pluripotent stem cells (iPSCs) derived from individuals with these mutations exhibited abnormal cytokinesis with delayed mitosis, multipolar spindles, and increased apoptosis, rescued by CRISPR/Cas9 genome editing. Our results highlight the importance of cytokinesis in the pathology of primary microcephaly.


Subject(s)
Alleles , Cytokinesis/genetics , Intracellular Signaling Peptides and Proteins/genetics , Microcephaly/genetics , Microcephaly/pathology , Mitosis/genetics , Mutation, Missense/genetics , Protein Serine-Threonine Kinases/genetics , Apoptosis/genetics , Centrosome/metabolism , Child , Child, Preschool , Female , Genes, Recessive , Humans , Infant, Newborn , Male , Pedigree
7.
J Biol Chem ; 288(2): 1295-306, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23155049

ABSTRACT

Accumulation of ß-amyloid (Aß) deposits is a primary pathological feature of Alzheimer disease that is correlated with neurotoxicity and cognitive decline. The role of glycogen synthase kinase-3 (GSK-3) in Alzheimer disease pathogenesis has been debated. To study the role of GSK-3 in Aß pathology, we used 5XFAD mice co-expressing mutated amyloid precursor protein and presenilin-1 that develop massive cerebral Aß loads. Both GSK-3 isozymes (α/ß) were hyperactive in this model. Nasal treatment of 5XFAD mice with a novel substrate competitive GSK-3 inhibitor, L803-mts, reduced Aß deposits and ameliorated cognitive deficits. Analyses of 5XFAD hemi-brain samples indicated that L803-mts restored the activity of mammalian target of rapamycin (mTOR) and inhibited autophagy. Lysosomal acidification was impaired in the 5XFAD brains as indicated by reduced cathepsin D activity and decreased N-glycoyslation of the vacuolar ATPase subunit V0a1, a modification required for lysosomal acidification. Treatment with L803-mts restored lysosomal acidification in 5XFAD brains. Studies in SH-SY5Y cells confirmed that GSK-3α and GSK-3ß impair lysosomal acidification and that treatment with L803-mts enhanced the acidic lysosomal pool as demonstrated in LysoTracker Red-stained cells. Furthermore, L803-mts restored impaired lysosomal acidification caused by dysfunctional presenilin-1. We provide evidence that mTOR is a target activated by GSK-3 but inhibited by impaired lysosomal acidification and elevation in amyloid precursor protein/Aß loads. Taken together, our data indicate that GSK-3 is a player in Aß pathology. Inhibition of GSK-3 restores lysosomal acidification that in turn enables clearance of Aß burdens and reactivation of mTOR. These changes facilitate amelioration in cognitive function.


Subject(s)
Acids/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/physiology , Disease Models, Animal , Glycogen Synthase Kinase 3/antagonists & inhibitors , Lysosomes/metabolism , TOR Serine-Threonine Kinases/metabolism , Alzheimer Disease/physiopathology , Animals , Autophagy , Brain/metabolism , Cell Line , Electrophoresis, Polyacrylamide Gel , Humans , In Vitro Techniques , Mice
8.
Ann Neurol ; 69(1): 170-80, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21280087

ABSTRACT

OBJECTIVE: The cleavage of amyloid precursor protein by γ-secretase is an important aspect of the pathogenesis of Alzheimer's disease. γ-Secretase also cleaves other membrane proteins (eg, Notch), which control cell development and homeostasis. Presenilin 1 and 2 are considered important determinants of the γ-secretase catalytic site. Our aim was to investigate whether γ-secretase can be important for microglial phagocytosis of Alzheimer's disease ß-amyloid. METHODS: We investigated the role of γ-secretase in microglia activity toward ß-amyloid phagocytosis in cell culture using γ-secretase inhibitors and small hairpin RNA and presenilin-deficient mice. RESULTS: We found that γ-secretase inhibitors impair microglial activity as measured in gene expression, protein levels, and migration ability, which resulted in a reduction of soluble ß-amyloid phagocytosis. Moreover, microglia deficient in presenilin 1 and 2 showed impairment in phagocytosis of soluble ß-amyloid. Dysfunction in the γ-secretase catalytic site led to an impairment in clearing insoluble ß-amyloid from brain sections taken from an Alzheimer's disease mouse model when compared to microglia from wild-type mice. INTERPRETATION: We suggest for the first time, a dual role for γ-secretase in Alzheimer's disease. One role is the cleavage of the amyloid precursor protein for pathologic ß-amyloid production and the other is to regulate microglia activity that is important for clearing neurotoxic ß-amyloid deposits. Further studies of γ-secretase-mediated cellular pathways in microglia may provide useful insights into the development of Alzheimer's disease and other neurodegenerative diseases, providing future avenues for therapeutic intervention.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Peptides/biosynthesis , Plaque, Amyloid/pathology , Presenilins/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cells, Cultured , Macrophages, Peritoneal/physiology , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microglia/physiology , Phagocytosis/physiology , Plaque, Amyloid/metabolism , Presenilins/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Transfection/methods
9.
Int Immunol ; 21(7): 817-29, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19515797

ABSTRACT

Myocardial ischemia with subsequent reperfusion (MI/R) can lead to significant myocardial damage. Ischemia initiates inflammation at the blood-microvascular endothelial cell interface and contributes significantly to both acute injury and repair of the damaged tissue. We have found that MI/R injury in mice is associated with a cellular immune response to troponin. Myocardial cells exclusively synthesize troponin and release the troponin into the bloodstream following injury. Mucosally administered proteins induce T cells that secrete anti-inflammatory cytokines such as IL-10 and transforming growth factor beta at the anatomical site where the protein localizes. We found that nasal administration of the three subunits of troponin (C, I and T isoforms), given prior to or 1 h following MI/R, decreased infarct size by 40% measured 24 h later. At 1.5 months following MI/R, there was a 50% reduction in infarct size and improvement in cardiac function as measured by echocardiography. Protection was associated with a reduction of cellular immunity to troponin. Immunohistochemistry demonstrated increased IL-10 and reduced IFN-gamma in the area surrounding the ischemic infarct following nasal troponin. Adoptive transfer of CD4+ T cells to mice from nasally troponin-treated mice 1 h after the MI/R decreased infarct size by 72%, whereas CD4+ T cells from IL-10-/- mice or nasally BSA-treated mice had no effect. Our results demonstrate that IL-10-secreting CD4+ T cells induced by nasal troponin reduce injury following MI/R. Modulation of cardiac inflammation by nasal troponin provides a novel treatment to decrease myocardial damage and enhance recovery after myocardial ischemia.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Interleukin-10/immunology , Myocardial Reperfusion Injury/prevention & control , Troponin/administration & dosage , Administration, Intranasal , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/drug effects , Disease Models, Animal , Female , Interferon-gamma/antagonists & inhibitors , Interleukin-10/agonists , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocardium/immunology , Myocardium/pathology , Protein Isoforms/immunology , Troponin/immunology , Vaccination
10.
Article in English | MEDLINE | ID: mdl-20706642

ABSTRACT

Toll-like receptors (TLRs) are known to be expressed by innate immune response cells and to play a critical role in their activation against foreign pathogens. It was recently suggested that TLRs have an important role in the crosstalk between neurons and glial cells in the central nervous system (CNS). TLR signaling was reported to be associated with a yin-yang effect in the CNS. While TLR signaling was linked to neurogenesis, it was also found to be involved in the pathogenesis of neurodegenerative diseases. This paper will focus on TLR signaling in glial cells in neurodegenerative diseases such as Alzheimer's disease, prion diseases, amyotrophic lateral sclerosis, and Parkinson's disease. Understanding the pattern of TLR signaling in the glial cells may lead to the identification of new targets for therapeutic application.


Subject(s)
Amyloidosis , Neurodegenerative Diseases , Neuroglia/metabolism , Signal Transduction/physiology , Toll-Like Receptors/metabolism , Amyloidosis/metabolism , Amyloidosis/pathology , Amyloidosis/therapy , Animals , Humans , Immunity, Innate/immunology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/therapy , Neurogenesis/physiology , Neurons/metabolism
11.
Sleep ; 43(2)2020 02 13.
Article in English | MEDLINE | ID: mdl-31553459

ABSTRACT

Increasing evidence highlight the involvement of immune cells in brain activity and its dysfunction. The brain's immune compartment is a dynamic ensemble of cells that can fluctuate even in naive animals. However, the dynamics and factors that can affect the composition of immune cells in the naive brain are largely unknown. Here, we examined whether acute sleep deprivation can affect the brain's immune compartment (parenchyma, meninges, and choroid plexus). Using high-dimensional mass cytometry analysis, we broadly characterized the effects of short-term sleep deprivation on the immune composition in the mouse brain. We found that after 6 h of sleep deprivation, there was a significant increase in the abundance of B cells in the brain compartment. This effect can be accounted for, at least in part, by the elevated expression of the migration-related receptor, CXCR5, on B cells and its ligand, cxcl13, in the meninges following sleep deprivation. Thus, our study reveals that short-term sleep deprivation affects the brain's immune compartment, offering a new insight into how sleep disorders can affect brain function and potentially contribute to neurodegeneration and neuroinflammation.


Subject(s)
Brain , Sleep Deprivation , Animals , B-Lymphocytes , Brain Mapping , Cell Movement , Mice , Sleep Deprivation/complications
12.
J Mol Neurosci ; 55(2): 430-6, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24994540

ABSTRACT

Low-level laser therapy (LLLT) has been used to treat inflammation, tissue healing, and repair processes. We recently reported that LLLT to the bone marrow (BM) led to proliferation of mesenchymal stem cells (MSCs) and their homing in the ischemic heart suggesting its role in regenerative medicine. The aim of the present study was to investigate the ability of LLLT to stimulate MSCs of autologous BM in order to affect neurological behavior and ß-amyloid burden in progressive stages of Alzheimer's disease (AD) mouse model. MSCs from wild-type mice stimulated with LLLT showed to increase their ability to maturate towards a monocyte lineage and to increase phagocytosis activity towards soluble amyloid beta (Aß). Furthermore, weekly LLLT to BM of AD mice for 2 months, starting at 4 months of age (progressive stage of AD), improved cognitive capacity and spatial learning, as compared to sham-treated AD mice. Histology revealed a significant reduction in Aß brain burden. Our results suggest the use of LLLT as a therapeutic application in progressive stages of AD and imply its role in mediating MSC therapy in brain amyloidogenic diseases.


Subject(s)
Alzheimer Disease/therapy , Low-Level Light Therapy , Amyloid beta-Peptides/metabolism , Animals , Cognition , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/metabolism , Phagocytosis
13.
Nat Commun ; 4: 2030, 2013.
Article in English | MEDLINE | ID: mdl-23799536

ABSTRACT

In Alzheimer's disease, soluble amyloid-ß causes synaptic dysfunction and neuronal loss. Receptors involved in clearance of soluble amyloid-ß are not known. Here we use short hairpin RNA screening and identify the scavenger receptor Scara1 as a receptor for soluble amyloid-ß expressed on myeloid cells. To determine the role of Scara1 in clearance of soluble amyloid-ß in vivo, we cross Scara1 null mice with PS1-APP mice, a mouse model of Alzheimer's disease, and generate PS1-APP-Scara1-deficient mice. Scara1 deficiency markedly accelerates Aß accumulation, leading to increased mortality. In contrast, pharmacological upregulation of Scara1 expression on mononuclear phagocytes increases Aß clearance. This approach is a potential treatment strategy for Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Disease Progression , Leukocytes, Mononuclear/metabolism , Phagocytes/metabolism , Scavenger Receptors, Class A/deficiency , Animals , CD36 Antigens/metabolism , Cysteine Endopeptidases/pharmacology , Drug Combinations , HEK293 Cells , Humans , Leukocytes, Mononuclear/drug effects , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Microglia/drug effects , Microglia/metabolism , Phagocytes/drug effects , Presenilin-1/metabolism , Proteolysis/drug effects , RNA, Small Interfering/metabolism , Scavenger Receptors, Class A/metabolism , Solubility , Survival Analysis , Up-Regulation/drug effects
14.
PLoS One ; 6(1): e16564, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21305046

ABSTRACT

An increasing body of evidence indicates that accumulation of soluble oligomeric assemblies of ß-amyloid polypeptide (Aß) play a key role in Alzheimer's disease (AD) pathology. Specifically, 56 kDa oligomeric species were shown to be correlated with impaired cognitive function in AD model mice. Several reports have documented the inhibition of Aß plaque formation by compounds from natural sources. Yet, evidence for the ability of common edible elements to modulate Aß oligomerization remains an unmet challenge. Here we identify a natural substance, based on cinnamon extract (CEppt), which markedly inhibits the formation of toxic Aß oligomers and prevents the toxicity of Aß on neuronal PC12 cells. When administered to an AD fly model, CEppt rectified their reduced longevity, fully recovered their locomotion defects and totally abolished tetrameric species of Aß in their brain. Furthermore, oral administration of CEppt to an aggressive AD transgenic mice model led to marked decrease in 56 kDa Aß oligomers, reduction of plaques and improvement in cognitive behavior. Our results present a novel prophylactic approach for inhibition of toxic oligomeric Aß species formation in AD through the utilization of a compound that is currently in use in human diet.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/drug effects , Cinnamomum zeylanicum , Cognition Disorders/drug therapy , Administration, Oral , Alzheimer Disease/drug therapy , Animals , Cognition Disorders/prevention & control , Diptera , Disease Models, Animal , Mice , Mice, Transgenic , Protein Multimerization/drug effects , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL