Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Mol Neurosci ; 74(2): 55, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38776015

ABSTRACT

The dysregulation of lipid metabolism has been strongly associated with Alzheimer's disease (AD) and has intricate connections with various aspects of disease progression, such as amyloidogenesis, bioenergetic deficit, oxidative stress, neuroinflammation, and myelin degeneration. Here, a comprehensive bioinformatic assessment was conducted on lipid metabolism genes in the brains and peripheral blood of AD-derived transcriptome datasets, characterizing the correlation between differentially expressed genes (DEGs) of lipid metabolism and disease pathologies, as well as immune cell preferences. Through the application of weighted gene co-expression network analysis (WGCNA), modules eigengenes related to lipid metabolism were pinpointed, and the examination of their molecular functions within biological processes, molecular pathways, and their associations with pathological phenotypes and molecular networks has been characterized. Analysis of biological networks indicates notable discrepancies in the expression patterns of the DEGs between neuronal and immune cells, as well as variations in cell type enrichments within both brain tissue and peripheral blood. Additionally, drugs targeting the DEGs from central and peripheral and a diagnostic model for hub genes from the blood were retrieved and assessed, some of which were shown to be useful for therapeutic and diagnostic. These results revealed the distinctive pattern of transcriptionally abnormal lipid metabolism in central, peripheral, and immune cell activation, providing valuable insight into lipid metabolism for diagnosing and guiding more effective treatment for AD.


Subject(s)
Alzheimer Disease , Lipid Metabolism , Transcriptome , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Humans , Lipid Metabolism/genetics , Brain/metabolism , Gene Regulatory Networks
2.
Cell Commun Signal ; 22(1): 31, 2024 01 12.
Article in English | MEDLINE | ID: mdl-38216911

ABSTRACT

The intracellular deposition and intercellular transmission of α-synuclein (α-syn) are shared pathological characteristics among neurodegenerative disorders collectively known as α-synucleinopathies, including Parkinson's disease (PD). Although the precise triggers of α-synucleinopathies remain unclear, recent findings indicate that disruption of microglial homeostasis contributes to the pathogenesis of PD. Microglia play a crucial role in maintaining optimal neuronal function by ensuring a homeostatic environment, but this function is disrupted during the progression of α-syn pathology. The involvement of microglia in the accumulation, uptake, and clearance of aggregated proteins is critical for managing disease spread and progression caused by α-syn pathology. This review summarizes current knowledge on the interrelationships between microglia and α-synucleinopathies, focusing on the remarkable ability of microglia to recognize and internalize extracellular α-syn through diverse pathways. Microglia process α-syn intracellularly and intercellularly to facilitate the α-syn neuronal aggregation and cell-to-cell propagation. The conformational state of α-synuclein distinctly influences microglial inflammation, which can affect peripheral immune cells such as macrophages and lymphocytes and may regulate the pathogenesis of α-synucleinopathies. We also discuss ongoing research efforts to identify potential therapeutic approaches targeting both α-syn accumulation and inflammation in PD. Video Abstract.


Subject(s)
Parkinson Disease , Synucleinopathies , Humans , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism , alpha-Synuclein/therapeutic use , Synucleinopathies/metabolism , Synucleinopathies/pathology , Microglia/metabolism , Inflammation/metabolism , Homeostasis
3.
J Vis Exp ; (192)2023 02 24.
Article in English | MEDLINE | ID: mdl-36912554

ABSTRACT

Drosophila melanogaster is an ideal model organism for studying various diseases due to its abundance of advanced genetic manipulation techniques and diverse behavioral features. Identifying behavioral deficiency in animal models is a crucial measure of disease severity, for example, in neurodegenerative diseases where patients often experience impairments in motor function. However, with the availability of various systems to track and assess motor deficits in fly models, such as drug-treated or transgenic individuals, an economical and user-friendly system for precise evaluation from multiple angles is still lacking. A method based on the AnimalTracker application programming interface (API) is developed here, which is compatible with the Fiji image processing program, to systematically evaluate the movement activities of both adult and larval individuals from recorded video, thus allowing for the analysis of their tracking behavior. This method requires only a high-definition camera and a computer peripheral hardware integration to record and analyze behavior, making it an affordable and effective approach for screening fly models with transgenic or environmental behavioral deficiencies. Examples of behavioral tests using pharmacologically treated flies are given to show how the techniques can detect behavioral changes in both adult flies and larvae in a highly repeatable manner.


Subject(s)
Drosophila melanogaster , Drosophila , Animals , Behavior, Animal , Locomotion , Animals, Genetically Modified
4.
Brain Behav Immun ; 97: 22-31, 2021 10.
Article in English | MEDLINE | ID: mdl-34022373

ABSTRACT

Inappropriate synaptic development has been proposed as a potential mechanism of neurodevelopmental disorders, including attention-deficit hyperactivity disorder (ADHD). Major histocompatibility complex class I (MHCI), an immunity-associated molecule expressed by neurons in the brain, regulates synaptic development; however, the involvement of MHCI in these disorders remains elusive. We evaluated whether functional MHCI deficiency induced by ß2m-/-Tap1-/- double-knockout in mice leads to abnormalities akin to those seen in neurodevelopmental disorders. We found that functional MHCI deficiency induced locomotor hyperactivity, motor impulsivity, and attention deficits, three major symptoms of ADHD. In contrast, these mice showed normal spatial learning, behavioral flexibility, social behavior, and sensorimotor integration. In the analysis of the dopamine system, upregulation of dopamine D1 receptor (D1R) expression in the nucleus accumbens and a greater locomotor response to D1R agonist SKF 81297 were found in the functional MHCI-deficient mice. Low-dose methylphenidate, used for the treatment of ADHD patients, alleviated the three behavioral symptoms and suppressed c-Fos expression in the D1R-expressing medium spiny neurons of the mice. These findings reveal an unexpected role of MHCI in three major symptoms of ADHD and may provide a novel landmark in the pathogenesis of ADHD.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Genes, MHC Class I , Methylphenidate , Receptors, Dopamine D1 , Animals , Attention Deficit Disorder with Hyperactivity/genetics , Dopamine , Humans , Mice , Receptors, Dopamine D1/genetics , Social Behavior
5.
Commun Biol ; 2: 424, 2019.
Article in English | MEDLINE | ID: mdl-31799427

ABSTRACT

Mitochondrial degeneration is considered one of the major causes of Parkinson's disease (PD). Improved mitochondrial functions are expected to be a promising therapeutic strategy for PD. In this study, we introduced a light-driven proton transporter, Delta-rhodopsin (dR), to Drosophila mitochondria, where the mitochondrial proton-motive force (Δp) and mitochondrial membrane potential are maintained in a light-dependent manner. The loss of the PD-associated mitochondrial gene CHCHD2 resulted in reduced ATP production, enhanced mitochondrial peroxide production and lower Ca2+-buffering activity in dopaminergic (DA) terminals in flies. These cellular defects were improved by the light-dependent activation of mitochondrion-targeted dR (mito-dR). Moreover, mito-dR reversed the pathology caused by the CHCHD2 deficiency to suppress α-synuclein aggregation, DA neuronal loss, and elevated lipid peroxidation in brain tissue, improving motor behaviors. This study suggests the enhancement of Δp by mito-dR as a therapeutic mechanism that ameliorates neurodegeneration by protecting mitochondrial functions.


Subject(s)
Light , Mitochondria/metabolism , Mitochondria/radiation effects , Mitochondrial Diseases/etiology , Mitochondrial Diseases/metabolism , Motor Activity , Parkinson Disease/etiology , Parkinson Disease/metabolism , Protons , Animals , Biomarkers , Disease Models, Animal , Disease Susceptibility , Dopaminergic Neurons/metabolism , Drosophila , Models, Biological , Oxidative Stress , Reactive Oxygen Species/metabolism , alpha-Synuclein/metabolism
6.
Hum Mol Genet ; 28(23): 3895-3911, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31600778

ABSTRACT

Mutations in CHCHD2 are linked to a familial, autosomal dominant form of Parkinson's disease (PD). The gene product may regulate mitochondrial respiratory function. However, whether mitochondrial dysfunction induced by CHCHD2 mutations further yields α-synuclein pathology is unclear. Here, we provide compelling genetic evidence that mitochondrial dysfunction induced by PD-linked CHCHD2 T61I mutation promotes α-synuclein aggregation using brain autopsy, induced pluripotent stem cells (iPSCs) and Drosophila genetics. An autopsy of an individual with CHCHD2 T61I revealed widespread Lewy pathology with both amyloid plaques and neurofibrillary tangles that appeared in the brain stem, limbic regions and neocortex. A prominent accumulation of sarkosyl-insoluble α-synuclein aggregates, the extent of which was comparable to that of a case with α-synuclein (SNCA) duplication, was observed in CHCHD2 T61I brain tissue. The prion-like activity and morphology of α-synuclein fibrils from the CHCHD2 T61I brain tissue were similar to those of fibrils from SNCA duplication and sporadic PD brain tissues. α-Synuclein insolubilization was reproduced in dopaminergic neuron cultures from CHCHD2 T61I iPSCs and Drosophila lacking the CHCHD2 ortholog or expressing the human CHCHD2 T61I. Moreover, the combination of ectopic α-synuclein expression and CHCHD2 null or T61I enhanced the toxicity in Drosophila dopaminergic neurons, altering the proteolysis pathways. Furthermore, CHCHD2 T61I lost its mitochondrial localization by α-synuclein in Drosophila. The mislocalization of CHCHD2 T61I was also observed in the patient brain. Our study suggests that CHCHD2 is a significant mitochondrial factor that determines α-synuclein stability in the etiology of PD.


Subject(s)
DNA-Binding Proteins/genetics , Loss of Function Mutation , Parkinson Disease/genetics , Transcription Factors/genetics , alpha-Synuclein/chemistry , Aged , Animals , Autopsy , Brain/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , Disease Models, Animal , Drosophila , Female , Humans , Male , Middle Aged , Mitochondria/metabolism , Neurons/cytology , Parkinson Disease/metabolism , Pedigree , Protein Aggregates , Protein Stability , Transcription Factors/metabolism
7.
Proc Natl Acad Sci U S A ; 116(41): 20689-20699, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31548400

ABSTRACT

Mutations in the iPLA2-VIA/PLA2G6 gene are responsible for PARK14-linked Parkinson's disease (PD) with α-synucleinopathy. However, it is unclear how iPLA2-VIA mutations lead to α-synuclein (α-Syn) aggregation and dopaminergic (DA) neurodegeneration. Here, we report that iPLA2-VIA-deficient Drosophila exhibits defects in neurotransmission during early developmental stages and progressive cell loss throughout the brain, including degeneration of the DA neurons. Lipid analysis of brain tissues reveals that the acyl-chain length of phospholipids is shortened by iPLA2-VIA loss, which causes endoplasmic reticulum (ER) stress through membrane lipid disequilibrium. The introduction of wild-type human iPLA2-VIA or the mitochondria-ER contact site-resident protein C19orf12 in iPLA2-VIA-deficient flies rescues the phenotypes associated with altered lipid composition, ER stress, and DA neurodegeneration, whereas the introduction of a disease-associated missense mutant, iPLA2-VIA A80T, fails to suppress these phenotypes. The acceleration of α-Syn aggregation by iPLA2-VIA loss is suppressed by the administration of linoleic acid, correcting the brain lipid composition. Our findings suggest that membrane remodeling by iPLA2-VIA is required for the survival of DA neurons and α-Syn stability.


Subject(s)
Brain/pathology , Cell Membrane/pathology , Dopaminergic Neurons/pathology , Drosophila Proteins/metabolism , Group X Phospholipases A2/metabolism , Nerve Degeneration/pathology , Parkinson Disease/pathology , alpha-Synuclein/chemistry , Animals , Animals, Genetically Modified , Brain/metabolism , Cell Membrane/metabolism , Dopaminergic Neurons/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster , Endoplasmic Reticulum Stress , Female , Group VI Phospholipases A2/genetics , Group VI Phospholipases A2/metabolism , Group X Phospholipases A2/genetics , Humans , Male , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nerve Degeneration/metabolism , Parkinson Disease/metabolism , Phospholipids/metabolism , Synaptic Transmission , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
8.
Int J Mol Sci ; 20(4)2019 Feb 20.
Article in English | MEDLINE | ID: mdl-30791515

ABSTRACT

Mutations of coiled-coil-helix-coiled-coil-helix domain containing 2 (CHCHD2) and 10 (CHCHD10) have been found to be linked to Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and/or frontotemporal lobe dementia (FTD). CHCHD2 and CHCHD10 proteins, which are homologous proteins with 54% identity in amino acid sequence, belong to the mitochondrial coiled-coil-helix-coiled-coil-helix (CHCH) domain protein family. A series of studies reveals that these twin proteins form a multimodal complex, producing a variety of pathophysiology by the disease-causing variants of these proteins. In this review, we summarize the present knowledge about the physiological and pathological roles of twin proteins, CHCHD2 and CHCHD10, in neurodegenerative diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/etiology , Frontotemporal Dementia/etiology , Mitochondrial Proteins/genetics , Parkinson Disease/etiology , Transcription Factors/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , DNA-Binding Proteins , Disease Susceptibility , Frontotemporal Dementia/metabolism , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Mutation , Parkinson Disease/metabolism , Protein Binding , Protein Transport , Signal Transduction , Structure-Activity Relationship , Transcription Factors/chemistry , Transcription Factors/metabolism
9.
Methods Mol Biol ; 1759: 47-57, 2018.
Article in English | MEDLINE | ID: mdl-28324489

ABSTRACT

Mitochondrial quality control is a key process in tissues with high energy demands, such as the brain and muscles. Recent studies using Drosophila have revealed that the genes responsible for familial forms of juvenile Parkinson's disease (PD), PINK1 and Parkin regulate mitochondrial function and motility. Cell biological analysis using mammalian cultured cells suggests that the dysregulation of mitophagy by PINK1 and Parkin leads to neurodegeneration in PD. In this chapter, we describe the methods to monitor mitochondrial morphology in the indirect flight muscles of adult Drosophila and Drosophila primary cultured neurons and the methods to analyze the motility of mitochondria in the axonal transport of living larval motor neurons.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Mitochondria/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Animals , Axonal Transport , Drosophila/genetics , Drosophila Proteins/genetics , Mitochondria/genetics , Mitochondria, Muscle/metabolism , Molecular Imaging , Neurons/metabolism , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics
10.
Nat Commun ; 8: 15500, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28589937

ABSTRACT

Mutations in CHCHD2 have been identified in some Parkinson's disease (PD) cases. To understand the physiological and pathological roles of CHCHD2, we manipulated the expression of CHCHD2 in Drosophila and mammalian cells. The loss of CHCHD2 in Drosophila causes abnormal matrix structures and impaired oxygen respiration in mitochondria, leading to oxidative stress, dopaminergic neuron loss and motor dysfunction with age. These PD-associated phenotypes are rescued by the overexpression of the translation inhibitor 4E-BP and by the introduction of human CHCHD2 but not its PD-associated mutants. CHCHD2 is upregulated by various mitochondrial stresses, including the destabilization of mitochondrial genomes and unfolded protein stress, in Drosophila. CHCHD2 binds to cytochrome c along with a member of the Bax inhibitor-1 superfamily, MICS1, and modulated cell death signalling, suggesting that CHCHD2 dynamically regulates the functions of cytochrome c in both oxidative phosphorylation and cell death in response to mitochondrial stress.


Subject(s)
Cytochromes c/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Parkinson Disease/pathology , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Survival , DNA-Binding Proteins , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Drosophila melanogaster/ultrastructure , Electron Transport , Flight, Animal/physiology , Humans , Male , Mice , Mitochondria/ultrastructure , Models, Biological , Muscles/ultrastructure , Mutation/genetics , Nerve Degeneration/pathology , Oxidative Phosphorylation , Oxidative Stress , Parkinson Disease/genetics , Phenotype , Protein Binding , Protein Stability , Signal Transduction , Stress, Physiological , Ubiquitin-Protein Ligases/metabolism , Up-Regulation
11.
Hum Mol Genet ; 26(15): 2933-2948, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28482024

ABSTRACT

Mutations of the retromer component Vps35 and endosomal kinase LRRK2 are linked to autosomal dominant forms of familial Parkinson's disease (PD). However, the physiological and pathological roles of Vps35 and LRRK2 in neuronal functions are poorly understood. Here, we demonstrated that the loss of Drosophila Vps35 (dVps35) affects synaptic vesicle recycling, dopaminergic synaptic release and sleep behavior associated with dopaminergic activity, which is rescued by the expression of wild-type dVps35 but not the PD-associated mutant dVps35 D647N. Drosophila LRRK2 dLRRK together with Rab5 and Rab11 is also implicated in synaptic vesicle recycling, and the manipulation of these activities improves the Vps35 synaptic phenotypes. These findings indicate that defects of synaptic vesicle recycling in which two late-onset PD genes, Vps35 and LRRK2, are involved could be key aspects of PD etiology.


Subject(s)
Drosophila Proteins/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Vesicular Transport Proteins/metabolism , Animals , Animals, Genetically Modified , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Endocytosis/genetics , Endocytosis/physiology , Endosomes/metabolism , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mutation , Parkinson Disease/etiology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Synaptic Transmission , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism , Vesicular Transport Proteins/genetics
13.
PLoS Genet ; 11(9): e1005503, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26355680

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2) is a key molecule in the pathogenesis of familial and idiopathic Parkinson's disease (PD). We have identified two novel LRRK2-associated proteins, a HECT-type ubiquitin ligase, HERC2, and an adaptor-like protein with six repeated Neuralized domains, NEURL4. LRRK2 binds to NEURL4 and HERC2 via the LRRK2 Ras of complex proteins (ROC) domain and NEURL4, respectively. HERC2 and NEURL4 link LRRK2 to the cellular vesicle transport pathway and Notch signaling, through which the LRRK2 complex promotes the recycling of the Notch ligand Delta-like 1 (Dll1)/Delta (Dl) through the modulation of endosomal trafficking. This process negatively regulates Notch signaling through cis-inhibition by stabilizing Dll1/Dl, which accelerates neural stem cell differentiation and modulates the function and survival of differentiated dopaminergic neurons. These effects are strengthened by the R1441G ROC domain-mutant of LRRK2. These findings suggest that the alteration of Notch signaling in mature neurons is a component of PD etiology linked to LRRK2.


Subject(s)
Endosomes/metabolism , Parkinson Disease/enzymology , Protein Serine-Threonine Kinases/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Carrier Proteins/metabolism , Dopamine/metabolism , Drosophila , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases
14.
PLoS One ; 9(9): e107099, 2014.
Article in English | MEDLINE | ID: mdl-25268136

ABSTRACT

Major histocompatibility complex class I (MHCI) molecules were recently identified as novel regulators of synaptic plasticity. These molecules are expressed in various brain areas, especially in regions undergoing activity-dependent synaptic plasticity, but their role in the nucleus accumbens (NAc) is unknown. In this study, we investigated the effects of genetic disruption of MHCI function, through deletion of ß2-microblobulin, which causes lack of cell surface expression of MHCI. First, we confirmed that MHCI molecules are expressed in the NAc core in wild-type mice. Second, we performed electrophysiological recordings with NAc core slices from wild-type and ß2-microglobulin knock-out mice lacking cell surface expression of MHCI. We found that low frequency stimulation induced long-term depression in wild-type but not knock-out mice, whereas high frequency stimulation induced long-term potentiation in both genotypes, with a larger magnitude in knock-out mice. Furthermore, we demonstrated that knock-out mice showed more persistent behavioral sensitization to cocaine, which is a NAc-related behavior. Using this model, we analyzed the density of total AMPA receptors and their subunits GluR1 and GluR2 in the NAc core, by SDS-digested freeze-fracture replica labeling. After repeated cocaine exposure, the density of GluR1 was increased, but there was no change in total AMPA receptors and GluR2 levels in wild-type mice. In contrast, following repeated cocaine exposure, increased densities of total AMPA receptors, GluR1 and GluR2 were observed in knock-out mice. These results indicate that functional deficiency of MHCI enhances synaptic potentiation, induced by electrical and pharmacological stimulation.


Subject(s)
Histocompatibility Antigens Class I/genetics , Nucleus Accumbens/physiopathology , Animals , Excitatory Postsynaptic Potentials , Histocompatibility Antigens Class I/metabolism , Long-Term Potentiation , Long-Term Synaptic Depression , Male , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity , Nucleus Accumbens/metabolism , Receptors, AMPA/metabolism
15.
Folia Parasitol (Praha) ; 56(4): 242-50, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20128236

ABSTRACT

Protective immunity against murine malaria infection depends largely on the establishment of effective Th1 immune response during the early stages of infection. Experimental data suggest that the death of Plasmodium yoelii 17XL (Py 17XL) susceptible BALB/c mice results from the suppression of Th1 immune response mediated by CD4+CD25+Foxp3+ regulatory T cells (Tregs). However, the mechanism by which Tregs regulate Th1 immune response is poorly understood. Since immunity is initiated by dendritic cells (DCs), we analysed DC responses to Py 17XL in control and Treg-depleted BALB/c mice. Myeloid DC proliferation, phenotypic maturation and interleukin-12 (IL-12) production were strongly inhibited in control BALB/c mice. In contrast, plasmacytoid DC proliferation and IL-10 production were strongly enhanced in control BALB/c mice. In-vivo depletion of Tregs resulted in significantly reversed inhibition of DC response, which may contribute to the establishment of Th1 immune response, indicating that Tregs contribute to the suppression of Th1 immune response during malaria. These findings suggest Tregs contribute to prevent Th1 immune response establishment during the early stage of Py 17XL infection by inhibiting DC response.


Subject(s)
Dendritic Cells/physiology , Forkhead Transcription Factors/metabolism , Malaria/immunology , T-Lymphocytes, Regulatory/physiology , Th1 Cells/physiology , Animals , Female , Forkhead Transcription Factors/genetics , Malaria/parasitology , Mice , Mice, Inbred BALB C , Plasmodium yoelii
SELECTION OF CITATIONS
SEARCH DETAIL
...