Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Publication year range
1.
Front Immunol ; 12: 633796, 2021.
Article in English | MEDLINE | ID: mdl-33841415

ABSTRACT

Triggering receptor expressed on myeloid cells-2 (TREM2) and colony-stimulating factor 1 receptor (CSF1R) are crucial molecules for microgliopathy, which is characterized by microglia dysfunction and has recently been proposed as the neuropathological hallmark of neurological disorders. TREM2 and CSF1R are receptors expressed primarily in microglia in the brain and modulate microglial activation and survival. They are thought to be in close physical proximity. However, whether there is a direct interaction between these receptors remains elusive. Moreover, the physiological role and mechanism of the interaction of TREM2 and CSF1R remain to be determined. Here, we found that TREM2 interacted with CSF1R based on a co-immunoprecipitation assay. Additionally, we found that CSF1R knockdown significantly reduced the survival of primary microglia and increased the Trem2 mRNA level. In contrast, CSF1R expression was increased in Trem2-deficient microglia. Interestingly, administration of CSF1, the ligand of CSF1R, partially restored the survival of Trem2-deficient microglia in vitro and in vivo. Furthermore, CSF1 ameliorated Aß plaques deposition in Trem2-/-; 5XFAD mouse brain. These findings provide solid evidence that TREM2 and CSF1R have intrinsic abilities to form complexes and mutually modulate their expression. These findings also indicate the potential role of CSF1 in therapeutic intervention in TREM2 variant-bearing patients with a high risk of Alzheimer's disease (AD).


Subject(s)
Cell Survival , Membrane Glycoproteins/metabolism , Microglia/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Immunologic/metabolism , Animals , Brain/pathology , Disease Models, Animal , Gene Knockout Techniques , HEK293 Cells , Humans , Immunoprecipitation , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Microglia/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology
2.
J Cell Mol Med ; 25(9): 4516-4521, 2021 05.
Article in English | MEDLINE | ID: mdl-33783963

ABSTRACT

Both Colony-stimulating factor 1 receptor (CSF1R) and triggering receptor expressed on myeloid cells-2 (TREM2) are trans-membrane receptors and are expressed in the brain primarily by microglia. Mutations in these two microglia-expressed genes associated with neurodegenerative disease have recently been grouped under the term "microgliopathy". Several literatures have indicated that CSF1R and TREM2 encounters a stepwise shedding and TREM2 variants impair or accelerate the processing. However, whether CSF1R variant affects the shedding of CSF1R remains elusive. Here, plasmids containing human CSF1R or TREM2 were transiently transfected into the human embryonic kidney (HEK) 293T cells. Using Western Blot and/or ELISA assay, we demonstrated that, similar to those of TREM2, an N-terminal fragment (NTF) shedding of CSF1R ectodomain and a subsequent C-terminal fragment (CTF) of CSF1R intra-membrane were generated by a disintegrin and metalloprotease (ADAM) family member and by γ-secretase, respectively. And the shedding was inhibited by treatment with Batimastat, an ADAM inhibitor, or DAPT or compound E, a γ-secretase inhibitor. Importantly, we show that the cleaved fragments, both extracellular domain and intracellular domain of a common disease associated I794T variant, were decreased significantly. Together, our studies demonstrate a stepwise approach of human CSF1R cleavage and contribute to understand the pathogenicity of CSF1R I794T variant in adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP). These studies also suggest that the cleaved ectodomain fragment released from CSF1R may be proposed as a diagnostic biomarker for ALSP.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Leukoencephalopathies/pathology , Membrane Glycoproteins/metabolism , Mutant Proteins/metabolism , Mutation , Proteolysis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Immunologic/metabolism , Amyloid Precursor Protein Secretases/genetics , HEK293 Cells , Humans , Leukoencephalopathies/genetics , Leukoencephalopathies/metabolism , Membrane Glycoproteins/genetics , Mutant Proteins/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Immunologic/genetics
3.
FASEB J ; 34(8): 10984-10997, 2020 08.
Article in English | MEDLINE | ID: mdl-32613609

ABSTRACT

Mutation of Triggering receptor expressed on myeloid cells 2 (TREM2) impairs the response of microglia to amyloid-ß (Aß) pathology in Alzheimer's disease (AD), although the mechanism governing TREM2-regulated microglia recruitment to Aß plaques remains unresolved. Here, we confirm that TREM2 mutation attenuates microglial migration. Then, using Trem2-/- mice and an R47H variant mouse model for AD generated for this study, we show that TREM2 deficiency or the AD-associated R47H mutation results in inhibition of FAK and Rac1/Cdc42-GTPase signaling critical for cell migration. Intriguingly, treatment with CN04, a Rac1/Cdc42-GTPase activator, partially enhances microglial migration in response to oligomeric Aß42 in Trem2-/- or R47H microglia both in vitro and in vivo. Our study shows that the dysfunction of microglial migration in the AD-associated TREM2 R47H variant is caused by FAK/Rac1/Cdc42 signaling disruption, and that activation of this signaling ameliorates impaired microglial migration response to Aß42 , suggesting a therapeutic target for R47H-bearing patients with high risk of AD.


Subject(s)
Amyloid beta-Peptides/genetics , Cell Movement/genetics , Focal Adhesion Kinase 1/genetics , GTP Phosphohydrolases/genetics , Microglia/pathology , Myeloid Cells/metabolism , Neuropeptides/genetics , Peptide Fragments/genetics , cdc42 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain/pathology , Cells, Cultured , Disease Models, Animal , Loss of Function Mutation/genetics , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Myeloid Cells/pathology , Signal Transduction/genetics
4.
J Alzheimers Dis ; 74(2): 535-544, 2020.
Article in English | MEDLINE | ID: mdl-32065797

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD) includes a large spectrum of neurodegenerative disorders. OBJECTIVE: To identify the relationship of ErbB4 mutation and ALS/FTD. METHODS: Here, we report an atypical case of frontal variant behavioral abnormalities at the initial stage, a stable plateau stage of 5 years, and paralysis involving both upper and lower motor neurons followed by progressive cognitive dysfunction at the advanced stage. The clinical findings suggested a diagnosis of ALS/FTD, and genetic testing revealed erb-b2 receptor tyrosine kinase 4 (ErbB4) heterozygous mutation (c.2136 T>G, p.I712M), identified in an ALS pedigree previously. We modeled mutant ErbB4 protein through the SWISS-MODEL Server, and speculated on the structural change caused by the mutation. We also identified that ErbB4 (I712M) mutation led to reduced auto-phosphorylation of ErbB4 upon neuregulin-1 (NRG1) stimulation. RESULTS: A functional analysis of ErbB4 mutation demonstrated an obviously decreased auto-phosphorylation of ErbB4 involving in the pathogenesis of ALS/FTD. CONCLUSION: We firstly found ErbB4 mutation to be identified in ALS/FTD.


Subject(s)
Amyotrophic Lateral Sclerosis/diagnostic imaging , Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/diagnostic imaging , Frontotemporal Dementia/genetics , Mutation/physiology , Neuregulin-1/genetics , Receptor, ErbB-4/genetics , Amino Acid Sequence , Amyotrophic Lateral Sclerosis/metabolism , Female , Frontotemporal Dementia/metabolism , Humans , Middle Aged , Neuregulin-1/chemistry , Neuregulin-1/metabolism , Pedigree , Protein Structure, Secondary , Receptor, ErbB-4/chemistry , Receptor, ErbB-4/metabolism , Signal Transduction/physiology
5.
Neuroimmunomodulation ; 26(2): 67-76, 2019.
Article in English | MEDLINE | ID: mdl-30928987

ABSTRACT

OBJECTIVE(S): Neuroinflammation is an important contributor to the development of seizures and epilepsy. Micro-RNA-155 (miR-155) plays a critical role in immunity and -inflammation. This study aims to explore the function of miR-155 and miR-155-mediated inflammation in epilepsy. METHODS: About 8-week-old male C57BL/6 mice were administered an intraperitoneal injection (i.p.) of kainic acid (KA) (15 mg/kg) or saline. The mice in the KA group developing acute seizure were further subjected to intracerebroventricular injection (i.c.v.) of antagomir negative control (NC) or miR-155 antagomir. Animal behavior was observed according to Racine's scale, and electroencephalographs were recorded. Primary microglia were cultured and treated with antagomir NC or antagomir. Whole-cell electrophysiological recording was conducted to detect the spontaneous EPSCs and IPSCs in the neurons treated with different conditioned medium from those microglia. miR-155 were detected by qRT-PCR in those models, as well as in the brain or blood from epileptic patients and healthy controls. RESULTS: miR-155 was abundantly expressed in glial cells compared with neurons, and its expression was markedly elevated in the brain of epilepsy patients and KA-induced seizure mice. Silencing miR-155 attenuated KA-induced seizure, abnormal electroencephalography, proinflammatory cytokine expression, and microglia morphology change. Moreover, conditioned media from KA-treated microglia impaired neuron excitability, whereas conditioned media from KA and miR-155 antagomir co-treated microglia had no such effects. Finally, miR-155 levels were significantly higher in the blood of epilepsy patients than those of healthy controls. CONCLUSION(S): These findings demonstrate that aberrant upregulation of miR-155 contributes to epileptogenesis through inducing microglia neuroinflammation.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , MicroRNAs/metabolism , Microglia/metabolism , Seizures/metabolism , Adult , Animals , Convulsants/toxicity , Epilepsy, Temporal Lobe/immunology , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Kainic Acid/toxicity , Male , Mice , Mice, Inbred C57BL , MicroRNAs/immunology , Microglia/immunology , Seizures/chemically induced , Seizures/immunology
6.
Front Aging Neurosci ; 10: 395, 2018.
Article in English | MEDLINE | ID: mdl-30532704

ABSTRACT

Alzheimer's disease (AD) is the leading cause of age-related dementia among the elderly population. Recent genetic studies have identified rare variants of the gene encoding the triggering receptor expressed on myeloid cells-2 (TREM2) as significant genetic risk factors in late-onset AD (LOAD). TREM2 is specifically expressed in brain microglia and modulates microglial functions in response to key AD pathologies such as amyloid-ß (Aß) plaques and tau tangles. In this review article, we discuss recent research progress in our understanding on the role of TREM2 in microglia and its relevance to AD pathologies. In addition, we discuss evidence describing new TREM2 ligands and the role of TREM2 signaling in microglial survival and energy metabolism. A comprehensive understanding of TREM2 function in the pathogenesis of AD offers a unique opportunity to explore the potential of this microglial receptor as an alternative target in AD therapy.

7.
Front Pharmacol ; 9: 734, 2018.
Article in English | MEDLINE | ID: mdl-30050437

ABSTRACT

Epilepsy is a common neurological disease with recurrent seizures and neurobehavioral comorbidities, including cognitive impairment and psychiatric disorders. Recent studies suggest that L-3-n-butylphthalide (NBP), an extract from the seeds of Apium graveolens Linn. (Chinese celery), ameliorates cognitive dysfunction in ischemia and/or Alzheimer's disease animal models. However, little is known about the role of NBP in epilepsy and the associated comorbidities. Here, using a pilocarpine-induced chronic epileptic mouse model, we found that NBP supplement not only alleviated seizure severity and abnormal electroencephalogram, but also rescued cognitive and emotional impairments in these epileptic mice. The possible underlying mechanisms may be associated with the protective role of NBP in reducing neuronal loss and in restoring the expression of neural synaptic proteins such as postsynaptic density protein 95 (PSD95) and glutamic acid decarboxylase 65/67 (GAD65/67). In addition, NBP treatment increased the transcription of neuroprotective factors, brain-derived neurotrophic factor and Klotho. These findings suggest that NBP treatment may be a potential strategy for ameliorating epileptogenesis and the comorbidities of cognitive and psychological impairments.

8.
Article in Chinese | MEDLINE | ID: mdl-21207845

ABSTRACT

AIM: To investigate how vasonatrin peptide (VNP) can attenuate the growth-promoting effect of hypoxia in cardiac fibroblasts cultured from neonatal rats. METHODS: The cultured cardiac fibroblasts were divided randomly into four groups: control group, hypoxia group, hypoxia + VNP group and hypoxia + 8-Bromo-cGMP group. The growth of cardiac myocytes was measured by the means of MTT method. The effect of VNP on the intracellular level of cGMP and PCNA were measured by the means of radioimmunoassay and immunohistochemistry stain respectively. RESULTS: Hypoxia (24 h) significantly increased the MTT A490nm value of cardiac fibroblasts (P < 0.05 vs control group). Both VNP (10(-7) mol/L) and 8-Bromo-cGMP (10(-3) mol/L) decreased MTT A490 nm value in cardiac fibroblast (P < 0.05 vs hypoxia group). VNP (10(-7) mol/L) increased the intracellular level of cGMP (P < 0.05 vs control and hypoxia group). Hypoxia (24 h) significantly increased the expression of proliferating cell nuclear antigen (PCNA) in cardiac myocytes (P < 0.05, vs control group), but VNP (10(-7) mol/L) decreased it. CONCLUSION: VNP can attenuate hypoxia-induced growth-promoting effect in cardiac fibroblasts which is associated with the changes of cGMP and PCNA.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Cyclic GMP/metabolism , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/drug effects , Proliferating Cell Nuclear Antigen/metabolism , Animals , Animals, Newborn , Cell Hypoxia , Cells, Cultured , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...