Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(5)2024 Mar 02.
Article in English | MEDLINE | ID: mdl-38474161

ABSTRACT

Obesity is a serious global health challenge, closely associated with numerous chronic conditions including type 2 diabetes. Anemarrhena asphodeloides Bunge (AA) known as Jimo has been used to address conditions associated with pathogenic heat such as wasting-thirst in Korean Medicine. Timosaponin A3 (TA3), a natural compound extracted from AA, has demonstrated potential therapeutic effects in various disease models. However, its effects on diabetes and obesity remain largely unexplored. We investigated the anti-obesity and anti-diabetic properties of TA3 using in vitro and in vivo models. TA3 treatment in NCI-H716 cells stimulated the secretion of glucagon-like peptide 1 (GLP-1) through the activation of phosphorylation of protein kinase A catalytic subunit (PKAc) and 5'-AMP-activated protein kinase (AMPK). In 3T3-L1 adipocytes, TA3 effectively inhibited lipid accumulation by regulating adipogenesis and lipogenesis. In a high-fat diet (HFD)-induced mice model, TA3 administration significantly reduced body weight gain and food intake. Furthermore, TA3 improved glucose tolerance, lipid profiles, and mitigated hepatic steatosis in HFD-fed mice. Histological analysis revealed that TA3 reduced the size of white adipocytes and inhibited adipose tissue generation. Notably, TA3 downregulated the expression of lipogenic factor, including fatty-acid synthase (FAS) and sterol regulatory element-binding protein 1c (SREBP1c), emphasizing its potential as an anti-obesity agent. These findings revealed that TA3 may be efficiently used as a natural compound for tackling obesity, diabetes, and associated metabolic disorders, providing a novel approach for therapeutic intervention.


Subject(s)
Anti-Obesity Agents , Diabetes Mellitus, Type 2 , Saponins , Animals , Mice , Obesity/metabolism , Steroids/pharmacology , Anti-Obesity Agents/pharmacology , Adipogenesis , AMP-Activated Protein Kinases/metabolism , Lipids/pharmacology , 3T3-L1 Cells , Diet, High-Fat , Mice, Inbred C57BL
2.
Mol Neurodegener ; 19(1): 25, 2024 Mar 16.
Article in English | MEDLINE | ID: mdl-38493185

ABSTRACT

Age-dependent accumulation of amyloid plaques in patients with sporadic Alzheimer's disease (AD) is associated with reduced amyloid clearance. Older microglia have a reduced ability to phagocytose amyloid, so phagocytosis of amyloid plaques by microglia could be regulated to prevent amyloid accumulation. Furthermore, considering the aging-related disruption of cell cycle machinery in old microglia, we hypothesize that regulating their cell cycle could rejuvenate them and enhance their ability to promote more efficient amyloid clearance. First, we used gene ontology analysis of microglia from young and old mice to identify differential expression of cyclin-dependent kinase inhibitor 2A (p16ink4a), a cell cycle factor related to aging. We found that p16ink4a expression was increased in microglia near amyloid plaques in brain tissue from patients with AD and 5XFAD mice, a model of AD. In BV2 microglia, small interfering RNA (siRNA)-mediated p16ink4a downregulation transformed microglia with enhanced amyloid phagocytic capacity through regulated the cell cycle and increased cell proliferation. To regulate microglial phagocytosis by gene transduction, we used poly (D,L-lactic-co-glycolic acid) (PLGA) nanoparticles, which predominantly target microglia, to deliver the siRNA and to control microglial reactivity. Nanoparticle-based delivery of p16ink4a siRNA reduced amyloid plaque formation and the number of aged microglia surrounding the plaque and reversed learning deterioration and spatial memory deficits. We propose that downregulation of p16ink4a in microglia is a promising strategy for the treatment of Alzheimer's disease.


Subject(s)
Alzheimer Disease , Aged , Animals , Humans , Mice , Alzheimer Disease/metabolism , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid/metabolism , RNA, Small Interfering
3.
J Cereb Blood Flow Metab ; 44(7): 1102-1116, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38388375

ABSTRACT

Astrocytes undergo disease-specific transcriptomic changes upon brain injury. However, phenotypic changes of astrocytes and their functions remain unclear after hemorrhagic stroke. Here we reported hemorrhagic stroke induced a group of inflammatory reactive astrocytes with high expression of Gfap and Vimentin, as well as inflammation-related genes lipocalin-2 (Lcn2), Complement component 3 (C3), and Serpina3n. In addition, we demonstrated that depletion of microglia but not macrophages inhibited the expression of inflammation-related genes in inflammatory reactive astrocytes. RNA sequencing showed that blood-brain barrier (BBB) disruption-related gene matrix metalloproteinase-3 (MMP3) was highly upregulated in inflammatory reactive astrocytes. Pharmacological inhibition of MMP3 in astrocytes or specific deletion of astrocytic MMP3 reduced BBB disruption and improved neurological outcomes of hemorrhagic stroke mice. Our study demonstrated that hemorrhagic stroke induced a group of inflammatory reactive astrocytes that were actively involved in disrupting BBB through MMP3, highlighting a specific group of inflammatory reactive astrocytes as a critical driver for BBB disruption in neurological diseases.


Subject(s)
Astrocytes , Blood-Brain Barrier , Hemorrhagic Stroke , Matrix Metalloproteinase 3 , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Astrocytes/metabolism , Astrocytes/pathology , Mice , Matrix Metalloproteinase 3/metabolism , Hemorrhagic Stroke/pathology , Hemorrhagic Stroke/metabolism , Male , Inflammation/metabolism , Inflammation/pathology , Complement C3/metabolism , Microglia/metabolism , Microglia/pathology , Mice, Inbred C57BL , Lipocalin-2/metabolism , Vimentin/metabolism
4.
Article in English | MEDLINE | ID: mdl-38346858

ABSTRACT

Astrocytes play an integral role in the development, maturation, and refinement of neuronal circuits. Astrocytes secrete proteins and lipids that instruct the formation of new synapses and induce the maturation of existing synapses. Through contact-mediated signaling, astrocytes can regulate the formation and state of synapses within their domain. Through phagocytosis, astrocytes participate in the elimination of excess synaptic connections. In this work, we will review key findings on the molecular mechanisms of astrocyte-synapse interaction with a focus on astrocyte-secreted factors, contact-mediated mechanisms, and synapse elimination. We will discuss this in the context of typical brain development and maintenance, as well as consider the consequences of dysfunction in these pathways in neurological disorders, highlighting a role for astrocytes in health and disease.

5.
Curr Opin Neurobiol ; 84: 102840, 2024 02.
Article in English | MEDLINE | ID: mdl-38290370

ABSTRACT

Astrocytes interact with various cell types, including neurons, vascular cells, microglia, and peripheral immune cells. These interactions are crucial for regulating normal brain functions as well as modulating neuroinflammation in pathological conditions. Recent transcriptomic and proteomic studies have identified critical molecules involved in astrocytic crosstalk with other cells, shedding light on their roles in maintaining brain homeostasis in both healthy and diseased conditions. Astrocytes perform these various roles through either direct or indirect physical associations with neuronal synapses and vasculature. Furthermore, astrocytes can communicate with other immune cells, such as microglia, T cells, and natural killer cells, through secreted molecules during neuroinflammation. In this review, we discuss the critical molecular basis of this astrocytic crosstalk and the underlying mechanisms of astrocyte communication with other cells. We propose that astrocytes function as a central hub in inter-connecting neurons, vasculatures, and immune cells in healthy and diseased brains.


Subject(s)
Astrocytes , Neuroinflammatory Diseases , Humans , Astrocytes/metabolism , Proteomics , Brain/physiology , Neurons/physiology , Microglia/metabolism
6.
Immunity ; 56(9): 2105-2120.e13, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37527657

ABSTRACT

Childhood neglect and/or abuse can induce mental health conditions with unknown mechanisms. Here, we identified stress hormones as strong inducers of astrocyte-mediated synapse phagocytosis. Using in vitro, in vivo, and human brain organoid experiments, we showed that stress hormones increased the expression of the Mertk phagocytic receptor in astrocytes through glucocorticoid receptor (GR). In post-natal mice, exposure to early social deprivation (ESD) specifically activated the GR-MERTK pathway in astrocytes, but not in microglia. The excitatory post-synaptic density in cortical regions was reduced in ESD mice, and there was an increase in the astrocytic engulfment of these synapses. The loss of excitatory synapses, abnormal neuronal network activities, and behavioral abnormalities in ESD mice were largely prevented by ablating GR or MERTK in astrocytes. Our work reveals the critical roles of astrocytic GR-MERTK activation in evoking stress-induced abnormal behaviors in mice, suggesting GR-MERTK signaling as a therapeutic target for stress-induced mental health conditions.


Subject(s)
Astrocytes , Phagocytosis , Stress, Psychological , Animals , Child , Humans , Mice , Astrocytes/metabolism , c-Mer Tyrosine Kinase/genetics , Hormones/metabolism , Synapses/metabolism , Stress, Psychological/metabolism
7.
Curr Opin Neurobiol ; 81: 102732, 2023 08.
Article in English | MEDLINE | ID: mdl-37247606

ABSTRACT

In the central nervous system, synaptic pruning, the removal of unnecessary synaptic contacts, is an essential process for proper circuit maturation in neurodevelopment as well as for synaptic homeostasis in the adult stage. Dysregulation of synaptic pruning can contribute to the initiation and progression of various mental disorders, such as schizophrenia and depression, as well as neurodegenerative diseases including Alzheimer's disease. In the past 15 years, pioneering works have demonstrated that different types of glial cells regulate the number of synapses by selectively eliminating them through phagocytic molecular machinery. Although a majority of findings have been focused on microglia, it is increasingly evident that astrocytes function as a critical player in activity-dependent synapse elimination in developing, adult, and diseased brains. In this review, we will discuss recent findings showing the mechanisms and physiological importance of astrocyte-mediated synapse elimination in controlling synapses and circuit homeostasis. We propose that astrocytes play dominant and non-redundant roles in eliminating synapses during the activity-dependent circuit remodeling processes that do not involve neuro-inflammation.


Subject(s)
Astrocytes , Synapses , Humans , Astrocytes/physiology , Synapses/physiology , Central Nervous System , Neuroglia , Phagocytosis
8.
Mol Ther ; 31(4): 1002-1016, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36755495

ABSTRACT

Fabry disease (FD), a lysosomal storage disorder, is caused by defective α-galactosidase (GLA) activity, which results in the accumulation of globotriaosylceramide (Gb3) in endothelial cells and leads to life-threatening complications such as left ventricular hypertrophy (LVH), renal failure, and stroke. Enzyme replacement therapy (ERT) results in Gb3 clearance; however, because of a short half-life in the body and the high immunogenicity of FD patients, ERT has a limited therapeutic effect, particularly in patients with late-onset disease or progressive complications. Because vascular endothelial cells (VECs) derived from FD-induced pluripotent stem cells display increased thrombospondin-1 (TSP1) expression and enhanced SMAD2 signaling, we screened for chemical compounds that could downregulate TSP1 and SMAD2 signaling. Fasudil reduced the levels of p-SMAD2 and TSP1 in FD-VECs and increased the expression of angiogenic factors. Furthermore, fasudil downregulated the endothelial-to-mesenchymal transition (EndMT) and mitochondrial function of FD-VECs. Oral administration of fasudil to FD mice alleviated several FD phenotypes, including LVH, renal fibrosis, anhidrosis, and heat insensitivity. Our findings demonstrate that fasudil is a novel candidate for FD therapy.


Subject(s)
Fabry Disease , Animals , Mice , Fabry Disease/drug therapy , Fabry Disease/genetics , Endothelial Cells/metabolism , alpha-Galactosidase/genetics , Phenotype , Enzyme Replacement Therapy
9.
Antioxidants (Basel) ; 12(2)2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36830049

ABSTRACT

Ischemic stroke is caused by insufficient blood flow to the brain. Astrocytes have a role in bidirectionally converting pyruvate, generated via glycolysis, into lactate and then supplying it to neurons through astrocyte-neuron lactate shuttle (ANLS). Pyruvate kinase M2 (PKM2) is an enzyme that dephosphorylates phosphoenolpyruvate to pyruvate during glycolysis in astrocytes. We hypothesized that a reduction in lactate supply in astrocyte PKM2 gene deletion exacerbates neuronal death. Mice harboring a PKM2 gene deletion were established by administering tamoxifen to Aldh1l1-CreERT2; PKM2f/f mice. Upon development of global cerebral ischemia, mice were immediately injected with sodium l-lactate (250 mg/kg, i.p.). To verify our hypothesis, we compared oxidative damage, microtubule disruption, ANLS disruption, and neuronal death between the gene deletion and control subjects. We observed that PKM2 gene deletion increases the degree of neuronal damage and impairment of lactate metabolism in the hippocampal region after GCI. The lactate administration groups showed significantly reduced neuronal death and increases in neuron survival and cognitive function. We found that lactate supply via the ANLS in astrocytes plays a crucial role in maintaining energy metabolism in neurons. Lactate administration may have potential as a therapeutic tool to prevent neuronal damage following ischemic stroke.

10.
Neural Regen Res ; 18(8): 1770-1776, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36751804

ABSTRACT

Recent studies have shown that microglia/macrophages and astrocytes can mediate synaptic phagocytosis through the MER proto-oncokinase in developmental or stroke models, but it is unclear whether the same mechanism is also active in traumatic brain injury. In this study, we established a mouse model of traumatic brain injury and found that both microglia/macrophages and astrocytes phagocytosed synapses and expression of the MER proto-oncokinase increased 14 days after injury. Specific knockout of MER in microglia/macrophages or astrocytes markedly reduced injury volume and greatly improved neurobehavioral function. In addition, in both microglia/macrophages-specific and astrocytes-specific MER knock-out mice, the number of microglia/macrophage and astrocyte phagocytosing synapses was markedly decreased, and the total number of dendritic spines was increased. Our study suggested that MER proto-oncokinase expression in microglia/macrophages and astrocytes may play an important role in synaptic phagocytosis, and inhibiting this process could be a new strategy for treating traumatic brain injury.

11.
Neurosci Res ; 187: 3-13, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36170922

ABSTRACT

Based on experience during our life, neuronal connectivity continuously changes through structural remodeling of synapses. Recent studies have shown that the complex interaction between astrocytes and synapses regulates structural synapse remodeling by inducing the formation and elimination of synapses, as well as their functional maturation. Defects in this astrocyte-mediated synapse remodeling cause problems in not only neuronal network activities but also animal behaviors. Moreover, in various neurological disorders, astrocytes have been shown to play central roles in the initiation and progression of synaptic pathophysiology through impaired interactions with synapses. In this review, we will discuss recent studies identifying the novel roles of astrocytes in neuronal circuit remodeling, focusing on synapse formation and elimination. We will also discuss the potential implication of defective astrocytic function in evoking various brain disorders.


Subject(s)
Brain Diseases , Nervous System Diseases , Animals , Astrocytes/physiology , Synapses/physiology , Neurons/physiology , Neuronal Plasticity/physiology
12.
Soc Work Public Health ; 38(5-8): 387-399, 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-38294156

ABSTRACT

In March, 2020, during the COVID-19 pandemic in Korea, the first Community Treatment Center (CTC), which is a motel-type Alternate Care Site (ACS) for mild and asymptomatic patients, was opened. This is a case study of the first Community treatment center prepared to respond to COVID-19. One of the researchers worked as a medical doctor in one of the CTCs operated by the Korean government. The CTC's eight medical staff members were interviewed in-depth one-on-one. Then the data obtained from observation, collection, and interview were triangulated. In this study, it was identified based on the 4S factor that evaluates the surge capacity to meet the medical needs of CTC. And how the CTC was operated from a medical and social welfare perspective and what problems appeared to patients during the operation were analyzed. Three dormitories of a national training center were used as the CTC. Each patient used a room equipped with a toilet, a shower, and a washbasin. Medical staff and government officials with various backgrounds were dispatched. Telemedicine was also used to prevent the spread of infection. The CTC made a significant contribution to both medical and social welfare fields. It provided patients psychological stability in a comfortable environment. But some patients had psychological problems and difficulties involving work and family care. Various efforts in conjunction with participation from social workers are required to reduce these problems.


Subject(s)
COVID-19 , Telemedicine , Humans , Surge Capacity , Pandemics , Republic of Korea
13.
Cell Rep ; 40(12): 111398, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36130507

ABSTRACT

Myelin transcription factor 1 like (Myt1l), a zinc-finger transcription factor, promotes neuronal differentiation and is implicated in autism spectrum disorder (ASD) and intellectual disability. However, it remains unclear whether Myt1l promotes neuronal differentiation in vivo and its deficiency in mice leads to disease-related phenotypes. Here, we report that Myt1l-heterozygous mutant (Myt1l-HT) mice display postnatal age-differential ASD-related phenotypes: newborn Myt1l-HT mice, with strong Myt1l expression, show ASD-like transcriptomic changes involving decreased synaptic gene expression and prefrontal excitatory synaptic transmission and altered righting reflex. Juvenile Myt1l-HT mice, with markedly decreased Myt1l expression, display reverse ASD-like transcriptomes, increased prefrontal excitatory transmission, and largely normal behaviors. Adult Myt1l-HT mice show ASD-like transcriptomes involving astrocytic and microglial gene upregulation, increased prefrontal inhibitory transmission, and behavioral deficits. Therefore, Myt1l haploinsufficiency leads to ASD-related phenotypes in newborn mice, which are temporarily normalized in juveniles but re-appear in adults, pointing to continuing phenotypic changes long after a marked decrease of Myt1l expression in juveniles.


Subject(s)
Autism Spectrum Disorder , Animals , Autism Spectrum Disorder/genetics , Disease Models, Animal , Mice , Nerve Tissue Proteins , Synaptic Transmission , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/genetics
14.
Nat Med ; 28(9): 1802-1812, 2022 09.
Article in English | MEDLINE | ID: mdl-35927581

ABSTRACT

Clearing amyloid-ß (Aß) through immunotherapy is one of the most promising therapeutic approaches to Alzheimer's disease (AD). Although several monoclonal antibodies against Aß have been shown to substantially reduce Aß burden in patients with AD, their effects on improving cognitive function remain marginal. In addition, a significant portion of patients treated with Aß-targeting antibodies experience brain edema and microhemorrhage associated with antibody-mediated Fc receptor activation in the brain. Here, we develop a phagocytosis inducer for Aß consisting of a single-chain variable fragment of an Aß-targeting monoclonal antibody fused with a truncated receptor binding domain of growth arrest-specific 6 (Gas6), a bridging molecule for the clearance of dead cells via TAM (TYRO3, AXL, and MERTK) receptors. This chimeric fusion protein (αAß-Gas6) selectively eliminates Aß plaques through TAM receptor-dependent phagocytosis without inducing NF-kB-mediated inflammatory responses or reactive gliosis. Furthermore, αAß-Gas6 can induce synergistic clearance of Aß by activating both microglial and astrocytic phagocytosis, resulting in better behavioral outcomes with substantially reduced synapse elimination and microhemorrhage in AD and cerebral amyloid angiopathy model mice compared with Aß antibody treatment. Our results suggest that αAß-Gas6 could be a novel immunotherapeutic agent for AD that overcomes the side effects of conventional antibody therapy.


Subject(s)
Alzheimer Disease , Single-Chain Antibodies , Alzheimer Disease/drug therapy , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Disease Models, Animal , Mice , Mice, Transgenic , NF-kappa B , Plaque, Amyloid/drug therapy , Receptors, Fc/therapeutic use , Single-Chain Antibodies/therapeutic use , c-Mer Tyrosine Kinase
15.
Cell Death Dis ; 13(7): 603, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35831271

ABSTRACT

Autophagy is a biological process that maintains cellular homeostasis and regulates the internal cellular environment. Hyperactivating autophagy to trigger cell death has been a suggested therapeutic strategy for cancer treatment. Mechanistic target of rapamycin (mTOR) is a crucial protein kinase that regulates autophagy; therefore, using a structure-based virtual screen analysis, we identified lomitapide, a cholesterol-lowering drug, as a potential mTOR complex 1 (mTORC1) inhibitor. Our results showed that lomitapide directly inhibits mTORC1 in vitro and induces autophagy-dependent cancer cell death by decreasing mTOR signaling, thereby inhibiting the downstream events associated with increased LC3 conversion in various cancer cells (e.g., HCT116 colorectal cancer cells) and tumor xenografts. Lomitapide also significantly suppresses the growth and viability along with elevated autophagy in patient-derived colorectal cancer organoids. Furthermore, a combination of lomitapide and immune checkpoint blocking antibodies synergistically inhibits tumor growth in murine MC38 or B16-F10 preclinical syngeneic tumor models. These results elucidate the direct, tumor-relevant immune-potentiating benefits of mTORC1 inhibition by lomitapide, which complement the current immune checkpoint blockade. This study highlights the potential repurposing of lomitapide as a new therapeutic option for cancer treatment.


Subject(s)
Antineoplastic Agents , Autophagic Cell Death , Colorectal Neoplasms , Animals , Antineoplastic Agents/pharmacology , Autophagy , Benzimidazoles , Cholesterol/pharmacology , Colorectal Neoplasms/drug therapy , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , TOR Serine-Threonine Kinases/metabolism
17.
Nat Aging ; 2(8): 726-741, 2022 08.
Article in English | MEDLINE | ID: mdl-37118130

ABSTRACT

The aging brain exhibits a region-specific reduction in synapse number and plasticity. Although astrocytes play central roles in regulating synapses, it is unclear how changes in astrocytes contribute to age-dependent cognitive decline and vulnerability to neurodegenerative diseases. Here, we identified a unique astrocyte subtype that exhibits dysregulated autophagy and morphology in aging hippocampus. In these autophagy-dysregulated astrocytes (APDAs), autophagosomes abnormally accumulate in swollen processes, impairing protein trafficking and secretion. We found that reduced mammalian target of rapamycin (mTOR) and proteasome activities with lysosomal dysfunction generate APDAs in an age-dependent manner. Secretion of synaptogenic molecules and astrocytic synapse elimination were significantly impaired in APDAs, suggesting that APDAs have lost their ability to control synapse number and homeostasis. Indeed, excitatory synapses and dendritic spines associated with APDAs were significantly reduced. Finally, we found that mouse brains with Alzheimer's disease showed a significantly accelerated increase in APDAs, suggesting potential roles for APDAs in age- and Alzheimer's disease-related cognitive decline and synaptic pathology.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/genetics , Astrocytes/metabolism , Proteostasis , Brain/pathology , Aging , Mammals
18.
J Cell Physiol ; 237(1): 128-148, 2022 01.
Article in English | MEDLINE | ID: mdl-34311499

ABSTRACT

Glucose metabolism is a mechanism by which energy is produced in form of adenosine triphosphate (ATP) by mitochondria and precursor metabolites are supplied to enable the ultimate enrichment of mature metabolites in the cell. Recently, glycolytic enzymes have been shown to have unconventional but important functions. Among these enzymes, pyruvate kinase M2 (PKM2) plays several roles including having conventional metabolic enzyme activity, and also being a transcriptional regulator and a protein kinase. Compared with the closely related PKM1, PKM2 is highly expressed in cancer cells and embryos, whereas PKM1 is dominant in mature, differentiated cells. Posttranslational modifications such as phosphorylation and acetylation of PKM2 change its cellular functions. In particular, PKM2 can translocate to the nucleus, where it regulates the transcription of many target genes. It is notable that PKM2 also acts as a protein kinase to phosphorylate several substrate proteins. Besides cancer cells and embryonic cells, astrocytes also highly express PKM2, which is crucial for lactate production via expression of lactate dehydrogenase A (LDHA), while mature neurons predominantly express PKM1. The lactate produced in cancer cells promotes tumor progress and that in astrocytes can be supplied to neurons and may act as a major source for neuronal ATP energy production. Thereby, we propose that PKM2 along with its different posttranslational modifications has specific purposes for a variety of cell types, performing unique functions.


Subject(s)
Leukemia, Myeloid, Acute , Pyruvate Kinase , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Glycolysis/physiology , Humans , Lactates , Protein Kinases/metabolism , Pyruvate Kinase/genetics
19.
Nat Commun ; 12(1): 6943, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34836962

ABSTRACT

The pathological role of reactive gliosis in CNS repair remains controversial. In this study, using murine ischemic and hemorrhagic stroke models, we demonstrated that microglia/macrophages and astrocytes are differentially involved in engulfing synapses in the reactive gliosis region. By specifically deleting MEGF10 and MERTK phagocytic receptors, we determined that inhibiting phagocytosis of microglia/macrophages or astrocytes in ischemic stroke improved neurobehavioral outcomes and attenuated brain damage. In hemorrhagic stroke, inhibiting phagocytosis of microglia/macrophages but not astrocytes improved neurobehavioral outcomes. Single-cell RNA sequencing revealed that phagocytosis related biological processes and pathways were downregulated in astrocytes of the hemorrhagic brain compared to the ischemic brain. Together, these findings suggest that reactive microgliosis and astrogliosis play individual roles in mediating synapse engulfment in pathologically distinct murine stroke models and preventing this process could rescue synapse loss.


Subject(s)
Brain/pathology , Gliosis/immunology , Infarction, Middle Cerebral Artery/complications , Synapses/pathology , Animals , Astrocytes/metabolism , Brain/cytology , Brain/immunology , Disease Models, Animal , Down-Regulation/immunology , Female , Gliosis/pathology , Humans , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Phagocytosis/genetics , Phagocytosis/immunology , RNA-Seq , Single-Cell Analysis , Synapses/immunology , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/metabolism
20.
EMBO J ; 40(15): e107121, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34013588

ABSTRACT

Glia contribute to synapse elimination through phagocytosis in the central nervous system. Despite the important roles of this process in development and neurological disorders, the identity and regulation of the "eat-me" signal that initiates glia-mediated phagocytosis of synapses has remained incompletely understood. Here, we generated conditional knockout mice with neuronal-specific deletion of the flippase chaperone Cdc50a, to induce stable exposure of phosphatidylserine, a well-known "eat-me" signal for apoptotic cells, on the neuronal outer membrane. Surprisingly, acute Cdc50a deletion in mature neurons causes preferential phosphatidylserine exposure in neuronal somas and specific loss of inhibitory post-synapses without effects on other synapses, resulting in abnormal excitability and seizures. Ablation of microglia or the deletion of microglial phagocytic receptor Mertk prevents the loss of inhibitory post-synapses and the seizure phenotype, indicating that microglial phagocytosis is responsible for inhibitory post-synapse elimination. Moreover, we found that phosphatidylserine is used for microglia-mediated pruning of inhibitory post-synapses in normal brains, suggesting that phosphatidylserine serves as a general "eat-me" signal for inhibitory post-synapse elimination.


Subject(s)
Microglia/metabolism , Phosphatidylserines/metabolism , Seizures/physiopathology , Synapses/physiology , c-Mer Tyrosine Kinase/metabolism , Animals , Brain/physiopathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phagocytosis/physiology , Seizures/genetics , c-Mer Tyrosine Kinase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...